Skip to main content

Full text of "USPTO Patents Application 09993647"

See other formats


(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) 



(19) World Intellectual Property Organization 

International Bureau 

(43) International Publication Date 
2 August 2007 (02.08.2007) 




PCT 



(10) International Publication Number 

wo 2007/087575 A2 



(51) International Patent Classification: 
A61K 31/192 (2006.01) 

(21) International Application Number: 

PCT/US2007/060995 

(22) International Filing Date: 24 January 2007 (24.01.2007) 



(25) Filing Language: 

(26) Publication Language: 



English 
English 



(30) Priority Data: 

60/761,612 
60/833,934 



24 January 2006 (24.0 1 .2006) US 
28 July 2006 (28.07.2006) US 



(63) Related by continuation (CON) or continuation-in-part 
(CIP) to earlier application: 

US 60/761,612 (CON) 

Filed on 24 January 2006 (24.01.2006) 

(71) Applicant (fo r all designated States except US)i UNIVER- 
SITY OF CfflCAGO [US/US]; 5801 S. Elhs, Chicago, IL 
60637 (US). 

(72) Inventors; and 

(75) Inventors/Applicants (for US only): MAITLAND, 
Mardi, Gomberg [US/US]; Chicago, IL (US). RATAIN, 
Mark [US/US]; 1040 West Oakdale Avenue, Chicago, IL 
60657 (US). GARCIA, Joe, GN [US/US] ; Chicago, IL 
(US). MAITLAND, Michael [US/US]; Chicago, IL (US). 
MORENO, Liliana [CO/US]; Chicago, IL (US). 



(74) Agent: LANDRUM, Charles, P.; Fulbright & Jaworski 
L.L.P., 600 Congress Avenue, Suite 2400, Austin, TX 
78701 (US). 

(81) Designated States (unless otherwise indicated, for every 
kind of national protection available): AE, AG, AL, AM, 
AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN, 
CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, 
GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, 
JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, 
LT, LU, LV, LY, MA, MD, MG, MK, MN, MW, MX, MY, 
MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, 
RU, SC, SD, SE, SG, SK, SL, SM, SV, SY, TJ, TM, TN, 
TR, TT, TZ, UA, UG, US (patent), UZ, VC, VN, ZA, ZM, 
ZW. 

(84) Designated States (unless otherwise indicated, for every 
kind of regional protection available): ARIPO (BW, GH, 
GM, KE, LS, MW, MZ, NA, SD, SL, SZ, TZ, UG, ZM, 
ZW), Eurasian (AM, AZ, BY, KG, KZ, MD, RU, TJ, TM), 
European (AT, BE, BG, CH, CY, CZ, DE, DK, EE, ES, FI, 
FR, GB, GR, HU, IE, IS, IT, LT, LU, LV, MC, NL, PL, PT, 
RO, SE, SI, SK, TR), OAPI (BE, BJ, CF, CG, CI, CM, GA, 
GN, GQ, GW, ML, MR, NE, SN, TD, TG). 

Published: 

— without international search report and to be republished 
upon receipt of that report 

For two-letter codes and other abbreviations, refer to the "Guid- 
ance Notes on Codes and Abbreviations" appearing at the begin- 
ning of each regular issue of the PCT Gazette. 



< 

IT) 

in 

00 



<S| (54) Title: COMPOSITIONS AND METHODS FOR TREATING PULMONARY HYPERTENSION 



O 



(57) Abstract: Compositions and methods of the invention are related to treating pulmonary hypertension using a Raf kinase in- 
hibitor, such as sorafenib. IQ a particular aspect, pulmonary hypertension is pulmonary arterial hypertension. 



wo 2007/087575 



PCT/US2007/060995 



DESCRIPTION 

COMPOSITIONS AND METHODS FOR TREATING PULMONARY 

HYPERTENSION 

BACKGROUND OF THE INVENTION 

This application claims priority to U.S. Provisional Patent applications serial number 
60/761,612 filed January 24, 2006 and serial number 60/833,934 filed July 28, 2006, each of 
which is incorporated herein by reference in its entirety. 

I. FIELD OF THE INVENTION 

The invention described herein is related generally to medicine and particularly 
cardiac and puhnonary medicine. The invention is fiirther related to therapeutic and 
prophylactic treatment of pulmonary hypertension and particularly pulmonary arterial 
hypertension. 

IT BACKGROUND 

Pulmonary hypertension ("PH") refers to a disease characterized by sustained 
elevations of puhnonary artery pressure (Rubin, 1997). Generally, a patient having a mean 
pulmonary artery pressirre equal to or greater than 25 mm Hg with a pulmonary capillary or 
left atrial pressure equal to or less than 15 mm Hg is characterized as having PH or as 
symptomatic of PH. These parameters may be measured in the subject at rest by right-heart 
catheterization. Pulmonary arterial hypertension ("PAH") includes idiopathic pulmonary 
arterial hypertension; familial pulmonary arterial hypertension; pulmonary arterial 
hypertension in the setting of connective tissue diseases (e,g., locahzed cutaneous systemic 
sclerosis (CREST syndrome), diffuse scleroderma, systemic lupus er3^ematosus, mixed 
connective tissue disease, and other less conmion diseases), portal hypertension, congenital 
left-to-right intracardiac shunts, and infection with the human immunodeficiency virus); and 
persistent pulmonary hypertension of the newborn. For a review of the mechanisms of 
disease related to PAH see Farber and Loscalso (2004), which is incorporated herein by 
reference in its entirety. 

The World Health Organization (WHO) has classified pulmonary hypertension into 
groups based on known causes. WHO group I includes patients with PAH including those 



1 



wo 2007/087575 



PCT/US2007/060995 



patients with Idiopathic PAH; FamiUal PAH, and Associated PAH, wliich is related to certain 
conditions including connective tissue diseases, congenital systemic-to-pulmonary-shunts, 
portal hypertension^ HIV infection, dnigs and toxins, glycogen storage disease, Gaucher's 
disease, hereditary hemorrhagic telangiectasia, hemoglobinopathies, myeloproliferative 
5 disorders, splenectomy, and others; PAH associated with significant venous or capillary 
involvement; and persistent pulmonary hypertension of the newborn. WHO group II includes 
patients with pulmonary venous hypertension. WHO group III includes patients with 
pulmonary hypertension associated with hypoxemia. WHO group IV includes patients with 
pulmonary hypertension due to chronic thrombotic disease, embolic disease or both. Finally, 
10 WHO group V includes patients with pulmonary hypertension due a variety of miscellaneous 
conditions. 

The WHO also classifies pulmonary hypertension into functional groups based on 
their exercise capacity and symptoms. WHO class I includes patients with PAH without 
limitations of physical activity. WHO class II includes patients with PAH resulting in slight 
15 limitation of physical activity. WHO class III includes patients with PAH resulting in 
marked limitation in physical activity. WHO class IV includes patients with PAH that are 
unable engage in physical activity without maaifesting symptoms. 

As stated above, PH is also associated with disorders of the respiratory system and/or 
hypoxemia, including chronic obstructive pulmonary disease^ interstitial lung disease, sleep- 
20 disordered breathing, alveolar hypoventilation disorders, chronic exposure to high altitude, 
neonatal lung disease and alveolar-capillary dysplasia (Humbert, 2004). PH is associated 
with chronic thrombotic disease, embolic disease or both, and with a variety of miscellaneous 
conditions. 

Based on the inadequate therapies available, a need remains for additional safe and 
25 effective methods of treating and managing PH. 

SUMMARY OF THE INVENTION 

The present invention includes the use of sorafenib, an inhibitor of multiple kinases 
important to angiogenesis (Raf-1, VEGFR-2, VEGFR-3, PDGFR-p) shown to have 
anticancer properties^ and sorafenib like compounds as an effective agent or therapy for 
30 reducing pulmonary arterial pressure or a symptom of pulmonary arterial pressure. The 
inventors contemplate using other inhibitors of angiogenesis or of VEGFR for reducing 



2 



wo 2007/087575 



PCT/US2007/060995 



pulmonary arterial pressiore or a syinptom of pulmonaay arterial pressure. The invention 
concerns therapeutic and preventative compositions and methods related to the various WHO 
groups of pulmonary arterial hypertension (PAH) (e.g,, group I that includes idiopathic 
pulmonary arterial hypertension (EPAH) and familial pulmonary arterial hypertension 
5 (FPAH)), and Associated PAH, with the others in WHO group I. In certain embodiments, 
PAH may be limited to a specific type of PAH. In certeiin embodiments the methods can 
include providing a VEGFR2 inhibitor (z.e., sorafenib like compounds) to a subject with 
pulmonary arterial hypertension (PAH)^ with symptoms of PAH^ or at risk for PAH, The 
VEGFR2 inliibitor can include abt-869, amg706, AZD2171, bay57-9352, sorafenib, XL647, 
10 XL999, GW786034, bevacizumab, PKC412, AEE788, PTK787 (vatalanib), OSI-930, OSI- 
817, SU11248, AG-013736, ZK3-4709, quinazoline ZD6474, pyrrolocarbazole CEP-7055, or 
CP-547632. 

The present invention, in some embodiments, concerns methods that include 
providing a kinase inhibitor, such as sorafenib, to a subject with pulmonary arterial 

15 hypertension (PAH), with symptoms of PAH, or at risk for PAH. The term "provide," and 
other related forms of the term, is used according to its ordinary meaning of to supply or to 
furnish, which may be accomplished directly or indirectly. It is contemplated that the patient 
may be provided sorafenib or other kinase inhibitor directly, such as by administering or 
prescribing, for example, sorafenib or a pharmaceutically acceptable salt thereof, or 

20 indirectly, such as by administering or prescribing a sorafenib prodrug or a pharmaceutically 
acceptable salt thereof, such that the subject is effectively provided with sorafenib. 

In some embodiments, the patient has a mean puhnonary artery pressure equal to or 
greater than 25 mm Hg with a pulmonary capillary or lefL atrial pressure equal to or less than 
15 nam Hg, and/or a pulmonary vascular resistance greater than or equal to 2 Wood Units. 

25 Pulmonary Vascular Resistance (PVR) is the general pressure load against which the right 
ventricle must pump to push blood through the lungs. PVR is typically expressed in Wood 
units which is defined by the formula: PVR = (MPAP minus PCWP) divided by cardiac 
output (CO). Less than 2 Wood Units equals the PVR of an average healthy person, z.e., 
MPAP is about 20 mmHg; LAP is about less than or equal to 15 mmHg; CO is about 5 liters 

30 per minute. MPAP is tiie Mean Average Pulmonary Artery Pressmre which is the average 
pressure in the pulmonary artery. MPAP can be calculated using the formula: [(2 times 
diastolic pulmonary artery pressure (DPAP) plus systolic pulmonary artery pressmre (SPAP)] 



3 



wo 2007/087575 



PCT/US2007/060995 



divided by 3, with the normal MPAP range tjApically being between 10 to 20 mmHg. PCWP 
is the Pulmonary Capillary Wedge Pressure which is an indirect measurement of pressure in 
the heart's left atrium which can be directly monitored during pulmonary catheterization. The 
normal PCWP range is typically between 5 to 15 mmHg. CO is the volume of blood ejected 
5 by the heart per minute and can be calculated by the formula: CO (in liters/minute) equals 
heart rate (in beats/minute) times stroke volume (in liters/beat). The normal CO range is 
between 4 to 8 L/min. Normal PVR is typically considered between 0.7 to 2.0 Wood units. 
These parameters may be measured in the subject at rest by right-heart catheterization. 

The term "administering" is used according to its ordinary meaning of to dispense, 
10 furnish, supply, or give, ha many embodiments of the invention, the subject may administer 
sorafenib to themselves or a medical practitioner may provide the sorafenib to the subject. 
The term "prescribing" is used according to its ordinary meaning of to advise or order the use 
of, and it is generally contemplated that a medical practitioner would prescribe the sorafenib 
to the subject, which the subject would then administer to themselves. It is contemplated that 
15 any embodiment in which sorafenib is provided to a subject may be implemented in the 
context of administering or prescribing sorafenib, a sorafenib prodrugs or a pharmaceutically 
acceptable salt thereof 

In further embodiments, it is contemplated that the subject is provided an amount of 
sorafenib that is effective to treat or prevent PAH. It is contemplated that the term '"treat" is 

20 used according to its ordinary meaning of to deal with a disease or affection, a part of the 
body, or a person in order to relieve or cure. Patients who have PAH or symptoms of PAH 
may be treated according to methods of the invention. It is further contemplated that 
treatment may include management of PAH or PAH symptoms, which means that treatment 
allows PAH or PAH symptoms to be controlled. The term ''prevent" is used according to its 

25 ordinary meaning of to preclude, stop, or hinder. It is contemplated that an "effective 
amoimf is an amount that achieves the stated goal, which may be treatment and/or 
prevention of PAH* It is contemplated that in the context of treatment an effective amount 
produces a therapeutic benefit^ which includes, but may not be necessarily limited to the 
following characteristics with respect to pulmonary arterial h3pertension: reducing mean 

30 pulmonary pressure, increasing cardiac output/cardiac index measured by either 
thermodilution or Fick, improving timed walk distance (e.g., six-minute walk), improviag 
metabolic equivalents (MET) (e.g-., exercise treadmill test), reducing anginal pain frequency. 



4 



wo 2007/087575 



PCT/US2007/060995 



reducing dyspnea, synocope, presyncope, symptoms of right heart failure including edema 
and ascites, preventing need for lung or heart transplant, reducing length of stay in intensive 
care, reducing length of stay in hospital, or prolonging life. 

Typically, a patient or subject is assessed by using the six-minute wallc test. The test 
5 is administered by preparing an unobstructed path of a Icnown distance, e.g., path of 50 feet 
(100 feet round-trip). A chair may be placed at each end of path. The patient is typically 
instructed to walk at his/her own pace and stop to rest if needed and to cover as much 
distance as possible. Typically, one would look for improvement between pretreatment 
assessment and post treatment assessments. The six minute walk distance may also be 
10 compared to distances walked by a comparable healthy population, typically by using 
equations from a published study of healthy people of the same age group, hnprovement in a 
patient's or subject's condition may be indicated by an increased six minute walk distance. 

Another method of assessing a patient or subject is the determination of tricuspid 
valve regurgitation velocity. Tricuspid valve regurgitation velocity is typically assessed by 

15 an echocardiogram, in particular Doppler echocardiograms. The trained sonographer 
possesses the requisite knowledge to carry out the echocardiograms in association with 
assessment by a qualified physician or technician for assessment. A typical 
echocardiographic imaging protocol includes a parasternal long-axis view; parasternal short- 
axis views at the aortic valve, mitral valve, and left ventricular levels; and apical 4-chamber, 

20 2-chamber, and long-axis views. Mitral, aortic, pulmonic and tricuspid valves can be imaged 
by color Doppler in multiple views to determine the degree of regurgitation. Measurements 
of various cardiac dimensions are performed according to American Society of 
Echocardiography convention. Pulmonary artery systolic pressure can be calculated utilizing 
a modified Bernoulli equation, PAP == 4v^4-RAP, where v = peak systolic velocity or tricuspid 

25 regurgitation jet recorded by continuous wave Doppler and RAP (right atrial pressure) is 
assumed to be 10 mmHg. Visual estimates of left ventricular ejection fraction can be made 
by integrating information from all views and left ventricular ejection fraction is considered 
normal if above 0.50, Improvement in a patient's or subject's condition may be indicated by 
a decrease in tricuspid valve regurgitation, as well a upgrading in the WHO fimctional class 

30 designation. 

In certain embodiments, the subject's mean pulmonary artery pressure is reduced to 
less than 25 mm Hg It is specifically contemplated that the subject may experience a 



5 



wo 2007/087575 



PCT/US2007/060995 



reduction in his mean pulmonary artery pressure of about, at least about, or at most about 1, 
2, 3, 4, 5, 10. 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75 or greater percent, or any range 
derivable therein, relative the value prior to treatment with sorafenib, therein, relative the 
value prior to treatment with sorafenib. Such measurements may be taken at rest using a 
right heart catheterization, though other methods may be used. 

Therefore, the present invention specifically includes methods for treating or 
preventing PAH in a patient comprising providing to the patient an effective amount of 
sorafenib or a pharmaceutically acceptable salt thereof Subjects and patients specifically 
include humans, in addition to other mammals. 

In some methods of the invention may fiirther include identifying a subject with PAH 
or symptoms of PAH or a patient at risk for PAH. This may be achieved by a number of ways 
knovm to those of skill in the art, including, but not necessarily limited to, taking a patient 
history, inquiring about family members with PAH, obtaining the level of pressure in the 
subject's pulmonary artery, identifying in the subject risk factors for PAH, assessing an 
electrocardiogram of the subject, assessing an echocardiogram of the subject, assessing 
pulmonary function tests (PFTs) of the patient, assessing a perfiision lung scan, a high 
resolution CT scan of the chest and assessing a right-heart cardiac catheterization with 
vasodilatory testing. 

In some embodiments of the invention, the subject has been diagnosed as having 
Class I, II, III, or IV PAH according to guidelines used for class diagnosis, such as by tlie 
New York Heart Association or as having Class I, II, III, or IV, such as by the World Health 
Organization. See Rubin, 2004, which is hereby incorporated by reference. The term "severe 
PAH" refers to Class 3 or 4 or Class III or IV, according to the relevant guidelines. 

In further embodiments of the invention, after the subject has taken or been given 
sorafenib or a sorafenib prodmg, he/she experiences a reduction in pulmonary pressure. In 
some methods of the invention the reduction is about, about at least or about at most a 
reduction of 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 percent, or any 
range derivable therein, relative the pressure prior to sorafenib intake. In other embodiments, 
there is a fall of mean pulmonary artery pressiu*e of at least about 10 ram Hg after treatment 
with sorafenib. It is contemplated that the fall may be about, at least about, or at most about. 



wo 2007/087575 



PCT/US2007/060995 



10, 15, 20, 25, 3O5 35, 40, 45, 50 mm Hg^ or any range derivable therein, after or during 
sorafenib treatment. 

In further embodiments, methods involve evaluating PAH in the subject before and/or 
after the subject has taken or been given sorafenib. This can be achieved by a number of 
5 ways that include, but are not necessarily limited to, having an electrocardiogram, an 
echocardiogram, pulmonary function tests (PFTs), a perfiasion lung scan, other vasodilator 
testing, and/or a right-heart cardiac catheterization. 

The present invention also relates to methods in which a subject is administered or 
prescribed multiple doses of sorafenib. The subject may take or be given 2, 3, 4, 5, 6, 7, 8, 9, 

10 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 100, 200, 300, 400, (twice 
daily- oncologic dose), 500 doses or more of sorafenib. It is also contemplated that a subject 
may be given or prescribed sorafenib indefinitely or for a set period of time, such as 1,2, 3, 4, 
5, 6, 7 days, 1, 2, 3, 4, 5, weeks, 1, 2, 3, 4, 5, 6, 7, 8, 9 10, 11, 12 months, and/or 1, 2, 3, 4, 5, 
6;, 7, 8, 9, 10 years or more, or any range derivable therein. Alternatively, it is contemplated 

15 that a patient continues to be prescribed or administered sorafenib until the subject can be 
classified as a Class 1 or 2 subject (or Class I or 11), the six mile walk distance is increased, 
the symptoms of PAH are relieved or reduced (such as triscuspid valve regurgitation), the 
mean pulmonary artery pressure is reduced to less than about 25 mm Hg or there is an 
improvement with respect to PAH or a symptom of PAH, such as a relative decrease or 

20 increase in the value of a measurement of about, at least about, or at most about 2, 3, 4, 5, 6, 
7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 76, 80, 
85, 90, 95, 100 or more percent or —fold, or any range derivable therein, as compared to a 
value of a measurement prior to or during sorafenib therapy. 

hi embodiments of the invention^ a single dose of sorafenib is between about 50 mg 
25 and about 400 or 600 mg twice daily of sorafenib, whereas a dose refers to a single, 
uninterrupted administration of sorafenib. A dose may be about, at least about, or at most 
about 0,1, 0.5, 1, 2, 3, 4, 5,-6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 
85, 90, 95, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 
500, 525, 550, 575, 600, 625, 650, 675, 700, 725, 750, 775, 800, 825/850, 875, 900, 925, 
30 950, 975, 1000 mg or mg/kg, or any range derivable therein. It is contemplated that a dosage 
of mg/kg refers to the mg amormt of sorafenib per kg of total body weight of the subject. It is 

7 



wo 2007/087575 



PCT/US2007/060995 



contemplated that when multiple doses are given to a patient^ the doses may vary in amount 
or they may be the same. 

Sorafenib may be introduced into a subject by a number of ways that include, but are 
not necessarily limited to, orally, intravenously, intraarterially, or inhalation. 

5 The present invention also covers methods that also include administering or 

prescribing at least a second PAH treatment (''secondary PAH treatment"). Other compounds 
or agents that have been used to treat PAH include, but are not limited to the following: an 
anticoagulant (such as Coumadin or Warfarin), an calcium channel blocker (such as 
amlodipine, diltiazem, nifedipine, felodipine, isradipine, nicardipine, or verapamil), a 

10 prostacyclin (such as epoprostenol, treprostinil, iloprost), nitric oxide (only used in acute 
settings), a diuretic, a cardiac glycoside (digoxin), an endothelin antagonist (including non- 
selective inhibition with bosentan), a phosphodiesterase inhibitor (such as sildenafil), an 
endopeptidase inhibitor, a lipid lowering agents a thromboxane inhibitor (such as terbogrel), 
or oxygen. It is contemplated that a combination of treatments that include sorafenib maybe 

15 employed including investigational agents such as sitaxentan and ambrisentan (selective 
endothelin antagonist), and tadalafil (long acting phosphodiesterase inhibitor). A description 
of different therapies is provided in Badesch et ah^ 2004, which is hereby incorporated by 
reference. 

In certain embodiments, the sorafenib or composition containing sorafenib, or a 
20 secondary PAH treatment may be administered or prescribed before, after, or during surgery. 
In some embodiments, the surgery is lung transplantation, in which case other treatment is 
not subsequently needed. 

Other embodiments of the invention include a pharmaceutical composition 
comprising sorafenib or a pharmaceutically acceptable salt thereof and at least a second PAH 
25 treatment, such as those discussed above. 

Any embodiment discussed with respect to one aspect of the invention applies to 
other aspects of the invention as well. It is further contemplated that embodiments discussed 
in the context of PAH may be applied in the context of PH, and vice versa. 

The embodiments in the Example section are understood to be embodiments of the 
30 invention that are applicable to all aspects of the invention. 

8 



wo 2007/087575 



PCT/US2007/060995 



The use of the term "or" in the claims is used to mean "and/or" unless explicitly 
indicated to refer to alternatives only or the alternatives are mutually exclusive^ although the 
disclosure supports a definition that refers to only alternatives and "and/or/' 

Throughout this application, the term "about" is used to indicate that a value includes 
5 the standard deviation of error for the device or method being employed to determine the 
value. 

Following long-standing patent law, the words "a" and "an/' when used in 
conjunction with the word "comprising" in the claims or specification, denotes one or more, 
unless specifically noted, 

10 Other objects, features and advantages of the present invention will become apparent 

from the following detailed description. It should be imderstood, however, that the detailed 
description and the specific examples, while indicating specific embodiments of the 
invention, are given by way of illustration only, since various changes and modifications 
within the spirit and scope of the invention will become apparent to those skilled in the art 

1 5 from this detailed description. 

DESCRIPTION OF THE DRAWINGS 

So the above-recited features, advantages, and objects of the invention, as well as 
others, will become clear and can be understood in detail, more particular descriptions and 
certain embodiments of the invention briefly summarized above are illustrated in the 
20 appended drawings. These drawings form a part of the specification. It is to be noted, 
however, that the appended drawings illustrate or are associated with certain embodiments of 
the invention and therefore are not to be considered limiting in their scope. 

FIGs. 1 A-IB FIG. lA Shows pulmonary arterial pressure (mm Hg) of Dahl Salt 
Sensitive (SS) rats under normoxic conditions, hypoxic conditions (10% FiOa), hypoxic 
25 conditions and SU5416 administration, hypoxic conditions and sorafenib administration, and 
hypoxic conditions and SU5416 and sorafenib administration. FIG. IB shows effects of 
Sorafenib on pulmonary artery pressures and remodeling in rodent PH. Sorafenib prevents 
hypoxia+Su5416 induced pulmonary HT and remodeling. 



9 



wo 2007/087575 



PCT/US2007/060995 



FIG. 2 Shows right ventricle systolic pressiire (mm Hg) of Dahl SS rats under 
normoxic conditions, hypoxic conditions, hypoxic conditions and SU5416 administration, 
hypoxic conditions and sorafenib administration, and hypoxic conditions and SU5416 and 
sorafenib administration. 

5 FIG. 3 Shows hematocrit % in Dahl SS rats under normoxic conditions, hypoxic 

conditions^ hypoxic conditions and SU5416 administration, hypoxic conditions and sorafenib 
administration, and hypoxic conditions and both SU5416 and sorafenib administration. 

FIGs, 4A-4B Show right ventricular/left ventricular 4- septum ratio values, in Dahl SS 
rats under normoxic conditions, hypoxic conditions, hypoxic conditions and SU5416 
10 administration, hypoxic conditions and sorafenib administration, and hypoxic conditions and 
SU5416 and sorafenib administration. Sorafenib prevented hypoxia+SU5416 development of 
right heart hypertrophy (RV/LV4-Septum) in Dahl SS rats, 

FIG. 5 Shows mean blood pressure (nmi Hg) of Dahl SS rats under normoxic 
conditions, hypoxic conditions, hypoxic conditions and SU5416 administration, hypoxic 
15 conditions and sorafenib admirustration, and hypoxic conditions and SU5416 and sorafenib 
administration. 

FIG. 6 Shows echocardiographic data on the calculated left ventricle mass (grams) of 
Dahl SS rats under normoxic conditions, hypoxic conditions, hypoxic conditions and SU5416 
administration, hypoxic conditions and sorafenib administration, and hypoxic conditions and 
20 SU5416 and sorafenib administration. 

FIG. 7 Shows echocardiographic data on the calculated aortic cardiac output (1/min) 
of Dahl SS rats under normoxic conditions, hypoxic conditions, hypoxic conditions and 
SU5416 administration, hypoxic conditions and sorafenib administration^ and hypoxic 
conditions and SU54I6 and sorafenib administration. 

25 FIG. 8 Shows echocardiographic data on the calculated pulmonary arterial pressure 

gradient (mm Hg) of Dahl SS rats under normoxic conditions, hypoxic conditions, hypoxic 
conditions and SU5416 administration, hypoxic conditions and sorafenib administration, and 
hypoxic conditions and SU5416 and sorafenib administration. 



10 



wo 2007/087575 



PCT/US2007/060995 



FIG. 9 Shows echocardiographic data on the calculated right ventricle free wall 
thickness of Dahl SS rats under normoxic conditions, hypoxic conditions, hypoxic conditions 
and SU5416 administration, hypoxic conditions and sorafenib administration^ and hypoxic 
conditions and SU5416 and sorafenib administration. 

5 FIG. 10 Shows echocardiographic data on the calculated pulmonary arterial pressure 

(mm Hg) of Dahl SS rats under normoxic conditions, hypoxic conditions, hypoxic conditions 
and SU5416 administration, hypoxic conditions and sorafenib administration, and hypoxic 
conditions and SU5416 and sorafenib administration. 

FIG. 1 1 Shows echocardiographic data on the calculated right ventricle thickness (cm) 
10 of Dahl SS rats under normoxic conditions, hypoxic conditions, hypoxic conditions and 
SU5416 administration, hypoxic conditions and sorafenib administration, and hypoxic 
conditions and SU5416 and sorafenib administration. 

FIGs. 12A-12B Show VEGFR-2 immuno staining of endothelial cells (arrows) 
occluding the lumen of a small pulmonary artery of SU5416 treated lung (rat) exposed to 
15 chronic hypoxia for 3 weeks (600X) (FIG. 12 A). Smooth Muscle Cell alpha actin 
immuno staining of a patent intra-alveoloar pulmonary artery of SU5416 treated lung (rat) 
exposed to chronic hypoxia for 3 weeks. Note that the precapillary vessel has acquired a 
well-defined medial smooth muscle cell layer (600X) (FIG. I2B). 

FIG. 13 Shows human pulmonary arteries demonstrating medial hypertrophy, intimal 
20 thickening, neomuscularization, and thrombin deposition (clot). 

FIG. 14 Shows a time course of clinical status, exercise, and hemodynamic before and 
after initiation of imatinib treatment. The figure demonstrates the improvement in functional 
class 6MW distance and decrease in pulmonary vascular resistance (patient had a decrease in 
both mean PAP and an increase in cardiac index). (Ghofrard et aL^ 2005) 

25 FIGs. 15A-15B Representative photomicrographs of rat lung at 21 days. 

Hematoxylin +-Eosin stained (H&E) sections (1 OX). A. Hypoxia + SU54 16 and B. Hypoxia 
4- SU5416 + Sorafenib. 

FIG. 16 Shows histopathology of rat lung under normoxia. Hypoxia, Hypoxia + 
SU5416+ Sorafenib, Hypoxia + Sorafenib, and Hypoxia + SU5416. The sections show 



11 



wo 2007/087575 



PCT/US2007/060995 



remodeling of the lung vasculature in injiired animals (Hypoxia-i-SU5416) and a dramatic 
improvement in rats that received daily sorafenib (Hypoxia -h SU5416+ Sorafenib), almost to 
Normoxia controls 

FIG. 17 Shows the Heath-Edwards grading of animals under normoxia. Hypoxia;, 
5 Hypoxia -I- SU5416 + Soarfenib, Hypoxia + Sorafenib, and Hypoxia + SU5416. The Heafli- 
Edwards grading system grades pulmonary hypertension by microscopic features. The 
general classification is as follows: 

Potentially Reversible ~ grade I Hypertrophy of the media of muscular pulmonary 
arteries. Extension of muscle into the wall of pulmonary arterioles; grade II Muscle 
10 . hypertrophy plus proliferation of intimal cells in arterioles and small muscular arteries; grade 
III Muscle hypertrophy plus subendothelial fibrosis. Eventually, concentric masses of fibrous 
tissue and reduplicated internal elastic lamina occlude the vascular lumen of arterioles and 
small muscular arteries. Large elastic arteries show atherosclerosis 

Usually Irreversible — grade IV Muscle hypertrophy is less apparent; progressive 
15 dilatation of small arteries^ especially those near vessels with intimal fibrous occlusion. 
Plexiform lesions occur; grade V Plexiform and angiomatoid lesions plus intra-alveolar 
hemosiderin-fiUed macrophages; grade VI Necrotizing arteritis with thrombosis. Fibrinoid 
necrosis of the arterial wall with a transmural infiltrate of polymorphonuclear leukocytes and 
eosinophils, 

20 DETAILED DESCRIPTION OF THE INVENTION 

Pulmonary arterial hypertension is an angiogenic proliferative vasculopathy resulting 
from abnormal endothelial and smooth muscle cell interactions. Over time, the vasculopathy 
causes a narrowing of the pulmonary artery and its branches, resulting in right heart failure 
and death. Therapies are directed primarily at dilating the narrowing vessels, and include: 

25 prostacyclins (epoprostenol, treprostinil, and iloprost), endothelin receptor blockers 
(bosentan^ sitaxsentan and ambrisarten^ which is in the FDA approval process)^ and 
phosphodiesterase inhibitors (sildenafil and tadalafil (investigational). All currently FDA- 
approved therapies improve six minute walk distance, (average 30 meters) with minimal 
change in hemodynamic measurements and only epoprostenol has a proved survival benefit. 

30 Drugs that inhibit processes important to pathological blood vessel branching and growth, 
represent a new class of therapeutic agents for pulmonary arterial hypertension. 

12 



wo 2007/087575 



PCT/US2007/060995 



Sorafenib is a bi-aryl urea that inliibits Raf-1 kinase, a regulator of endothelial 
apoptosis, VEGFR-2, PDGFR-P, and VEGFR-3, growth factor receptors necessary for 
angiogenesis, affecting vascular permeability and vessel stabilization through pericyte 
recruitment and maturation. The inventors have designed and executed a three week study to 
5 assess the safety and therapeutic activity of sorafenib in a hypoxia-induced model of 
pulmonary hypertension in Dahl SS rats. A compound with a reportedly more limited 
spectrum of kinase inhibitory activity, SU5416, was previously demonstrated to exacerbate 
pulmonary hypertension in hypoxic rats. 

Pulmonary hypertension refers to elevated blood pressure in the pulmonary 
10 circulation. Pulmonary hypertension can be either primary or secondary to pulmonary or 
cardiac disease. Typically, the pulmonary blood pressure in humans suffering from 
pulmonary hypertension is greater than a mean pulmonary artery pressure of 25 mm Hg. The 
common symptoms of PAH include dyspnea, fatigue, weakness, chest pain, recurrent 
syncope^ seizures^ light-headedness, neurologic deficits, leg edema and palpitations (Rich, 
15 1987; The Merck Manual 1999). 

One embodiment of the invention encompasses methods of treating, pallating^ 
preventing, and/or managing PAH by administration to a patient in need of such treatment, 
prevention, or management a therapeutically or prophylactically effective amount of 
sorafenib, or a phannaceutically acceptable derivative or prodrug thereof. 

20 As used herein, and unless otherwise indicated, the terms "pulmonary arterial 

hypertension," "PAH" and pulmonary hypertension "PH" and related disorders include, but 
are not limited to: Idiopathic PAH; Familial PAH, and Associated PAH, which is related to 
certain conditions including connective tissue disease, congenital systemic-to-pulmonary- 
shunts, portal hypertension^ HIV infection, drags and toxins, glycogen storage disease, 

25 Gaucher' s disease, hereditary hemorrhagic telangiectasia, hemoglobinopathies, 
myeloproliferative disorders, splenectomy, and others; PAH associated with significant 
venous or capillary involvement; and persistent pulmonary hypertension of the newborn. It is 
contemplated that embodiments discussed in the context of PAH may be applied in the 
context of PH, and vice versa. 

30 The term "prodrug" as used in this application refers to a precursor or derivative form 

of a pharmaceutically active substance that is capable of being enzymatically or non- 
13 



wo 2007/087575 



PCT/US2007/060995 



enzyiTLatically activated or converted into the more active parent form. See, e.g., Wilman 
(1986) and Stella et aL (1985). In certain aspects^ a prodrug is less cytotoxic^ has a longer 
half-life, may be targeted to a particular tissue and/or is more stable than the parent drug. For 
example, an amide group of sorafenib can be derivatized to a hydroxyalkyl or 
5 acycloxym ethyl derivative or other chemically appropriate derivative. The hydroxyalkyl may 
then be converted non-enzymatically to render sorafenib. The acycloxymethyl group can be 
acted upon by an esterase, vv^hich is then followed by a non-enzymatic conversion to 
sorafenib. 

Another embodiment of the invention encompasses a method of treating, preventing 
10 and/or managing PH, which comprises administering or prescribing to a patient in need of 
such treatment, prevention and/or management a therapeutically or prophylactically effective 
amount of sorafenib, or a pharmaceutically acceptable salt or prodrug thereof, and a 
therapeutically or prophylactically effective amount of a second agent. 

Examples of second agents include^ but are not limited to^ anticoagulants ^ diuretics, 
15 cardiac glycosides, calcium chamiel blockers, vasodilators, prostacyclin analogues, 
endothelin antagonists, phosphodiesterase inhibitors, endopeptidase inhibitors, lipid lowering 
agents, thromboxane inhibitors, or other agents found, for example, in the Physician*s Desk 
Reference 2003. Second agents can be large molecules {e,g,^ proteins) or small molecules 
{e.g.^ synthetic inorganic, organometallic, or organic molecules). Examples of specific 
20 second agents include, but are not limited to, amlodipine, diltiazem, nifedipine, adenosine, 
epoprostenol (Flolan®), treprostinil (Remodulin®), bosentan (Tracleer®), warfarin, digoxin, 
nitric oxide, L-arginine, iloprost, betaprost, and sildenafil (Viagra®), 

Another embodiment of the invention encompasses a method of reversing, reducing 
or avoiding an adverse effect associated with the administration of a therapeutic used to treat 
25 PH, which comprises administeringor prescribing to a patient in need thereof a 
therapeutically or prophylactically effective amount of sorafenib, or a pharmaceutically 
acceptable salt or prodmg thereof, and an optional second agent. 

Procedures such as lung transplantation may be necessary to treat PH patients who 
have failed to respond to medical therapy. It is believed that the combined use of sorafenib 
30 and lung transplantation in a patient suffering from PH can be beneficial. It is believed that 
sorafenib can work in combination with transplantation therapy, reducing complications such 



14 



wo 2007/087575 



PCT/US2007/060995 



as chronic rejection and opportunistic infections associated with the transplantation. 
Therefore, this invention encompasses a method of treating or managing PH, which 
comprises administering to a patient (e.g.^ a human) sorafenib, or a pharmaceutically 
acceptable salt or prodrug thereof, before, during, or after transplaatation therapy. 

5 Certain aspects of the invention encompass pharmaceutical compositions that can be 

used in methods of the invention. Specific compositions comprise sorafenib., or a 
pharmaceutically acceptable salt or prodrug thereof, and an optional second agent. 

Also encompassed by the invention are single unit dosage forms of sorafenib, or a 
pharmaceutically acceptable salt or prodmg thereof. 

10 Other aspects of the invention can encompass kits which comprise sorafenib, or a 

pharmaceutically acceptable salt or prodrug thereof, and a second agent. For example, a kit 
may contain the compound of the invention, and calcium channel blockers, vasodilators, 
prostacyclin analogues, endothelin antagonists, phosphodiesterase inhibitors, endopeptidase 
inhibitors, lipid lowering agents, thromboxane inhibitors or other agents used to treat PAH 

15 patients. 

As part of the spectrum of sorafenib's pharmacologic activity overlaps with SU5416, 
a study was performed to determine whether sorafenib had similar effects or with additional 
mechanisms of inhibitory activity could counteract the effects of SU5416, Dahl Salt 
Sensitive rats were divided into 5 groups: normoxia + vehicle (normal, healthy control), 

20 hypoxia + SU5416 (positive control), hypoxia + sorafenib (safety comparison with SU5416), 
and hypoxia + sorafenib + SU5416 (therapeutic activity assessment). Echocardiograms were 
performed on all rats at the start of the study. Except for the nomioxia group, all rats were 
maintained in a hypoxia chamber with a partial pressure of oxygen of 10% FiOa for the 
duration of the study. Rats in the two SU5416 groups received one subcutaneous injection of 

25 SU5416 at the start of the study (20 mg/kg). Stock sorafenib solutions were prepared every 
three days, crushing and dissolving sorafenib tablets in EL/ethanol (50:50; Sigma Cremophor 
EL, 95% ethyl alcohol) at final concentration of 4 mg/mL, protected from light exposure and 
stored at room temperature. Final dosing solutions were prepared on the day of use by 
dilution of the stock solution to 1 mg/mL with water and administered by gavage to the rats 

30 daily. After 3 weeks, all rats had echocardiography and hemodynamic studies. Organ and 
blood specimens were obtained for further evaluation. Echocardiography images were 

15 



wo 2007/087575 



PCT/US2007/060995 



obtained with short axis views demonstrating RV wall thickness when comparing Hypoxia + 
SU5416 4- sorafenib; Hypoxia + SU5416; and Control- Normoxia. RV 3DE measurements 
were also determined (diameters + area). A— RV major diameter, B= RV annulus diastolic 
diameter & area, C= RV at 1/3 RV major diameter & area, D — RV at 2/3 RV major diameter 
5 & area. 

As has been previously described, rats given SU5416 developed pulmonary 
hypertension measured by elevated right ventricular and pulmonary artery pressures, 
echocardiographic changes, and elevated right ventricle/left ventricular weights. Rats 
exposed to hypoxia had mildly elevated pressures compared with normoxia and there was no 

10 significant change in pressures or weights in rats given hypoxia plus sorafenib, Sorafenib 
appears to have a beneficial effect on pulmonary hypertensive rats as rats in hypoxia^ plus 
SU5416, plus sorafenib had pressures and weights similar to normoxia. The small sample 
size did not allow for significant change in echocardiographic data but there appeared to be a • 
positive trend with this combination. Pathologic specimen results are pending. Based on 

15 these results the experiments were repeated with 18 rats distmbuted in the same 5 groups 
(only difference was 3 additional rats in the normoxia group control group). The results were 
reproducible. In the initial experiment two rats died during induction of anesthesia for 
echocardiographic analysis (after unblinding-normoxia, hypoxia/SU5416). In the subsequent 
experiment, one rat died on day zero (after imblinding- 1 hypoxia/sorafenib on day three). 

20 These data suggest that sorafenib is a safe, therapeutic agent for the treatment of human 
pulmonary hypertension. Below are the results of the 2 experiments as one cohort. Also see 
FIGs. 1-1 L 

Table 1. Pressures derived firom SU5416+sorafenib study - 3 1/2 Wk Hypoxia + Sorafenib 
(Daily 2.5 mg/Kg) SU5416 (day zero 20 mg/kg i.p.) 





PA mmHg 
(+/-) SE 


RVSPmmHg 

(+/-) SE 


RV/LV kg 
SE 


BPmmHg 
{+/-) SE 


N 


Normoxia Vehicle 


12+/-2 


11+/-3 


0.173+/-0.007 


82+/-8 


7 


Hypoxia 


22+/-1 


20+/-2 


0,262+/-0.023 


94+/-4 


6 


Hypoxia-i-S U541 6 


33+/-3 


30+/-4 


0.389+/- 0-02 


78+/-4 


5 


Hypoxia +Sorafenib 


1 S+/-2 


1 1+1-2 


0.248+^ 0.02 


97+/-4 


6 


Hypoxia +Sorafenib+ 
SU5416 


19+/-2 


1 7+/-3 


0.202+/- 0.015 


108+/-10 


5 



25 

Sorafenib appears to protect SU5416-treated hypoxia-exposed rats from pulmonary 
arterial hypertension. Furthermore^ while sorafenib and SU5416 share some pharmacological 



16 



wo 2007/087575 



PCT/US2007/060995 



activity^ unlike SU5416 sorafenib does not exacerbate hypoxia-induced hypertension. These 
data suggest that sorafenib is safe and a potential therapeutic agent for the treatment of 
human pulmonary hypertension. 

III. COMPOUNDS OF THE INVENTION 

5 Sorafenib is a small molecule inhibitor of Raf kinase and Raf associated signaling 

pathways, including extracellular regulated kinases (ERKs). Sorafenib is also known as BAY 
43-9005^ its anti-cancer properties are described in US Patent Publication 20030125359, 
which is incorporated herein by reference. Sorafenib is known to target both the 
Raf/MEK/ERK signaling pathway inhibiting cell proliferation and the VEGFR-2/PDGFR-P 
10 pathway inhibiting angiogenesis. Sorafenib is currently being tested in the United States by 
Bayer/Onyx in phase III clinical trials for advanced renal cell carcinoma. Methods for the 
preparation of sorafenib and its related compounds is described in U.S. Patent Publication 
number 2001/0027202 and 2003/0139605, which are incorporated herein by reference in their 
entirety. 

1 5 A. Tyrosine kinase / VEGFR inhibitors 

A number of anti-cancer therapeutics are classified as tyrosine kinase inhibitors (for a 
review see Levitski and Mishani, 2006, which is incorporated herein by reference in its 
entirety). Tyrosine kinases include a number of cell surface receptors such as the VEGF 
receptors. VEGF is one of the key regulators for both physiological and pathological 
20 angiogenesis- Because of the multitude of cellular responses that are initiated and regulated 
by VEGF^ and because of its specificity for the vascular endothelium, VEGF takes an 
exceptional position among other growth factors (Petrova et ah Exp Cell Res, 253:117-130, 
1999). 

There are three VEGF-receptors (VEGFRl, VEGFR2, and VEGFR3). VEGFRl is 
25 mainly expressed in hematopoietic stem cells, macrophages and monocytes as well as in 
vascular endothelium. VEGFR2 is more characteristic of the vascular and lymphatic 
endothelium, whereas VEGFR3 is predominantly expressed in lymphatic endothelium (Cross 
et al, 2003). 

Since VEGFR2 is generally considered the most important transducer of VEGF- 
30 dependent angiogenesis, this receptor represents a major target within the angiogenesis- 

17 



wo 2007/087575 



PCT/US2007/060995 



related kinases. VEGFR2 inhibitors include^ but are not limited to abt-869, amg706, 
AZD2171, bay57-9352, bay43-9006 (sorafenib), XL647, XL999, GW786034, bevaciziimab, 
PKC412, AEE788, PTK787 (vatalanib), OSI-930, OSI-817, SU11248, AG-013736, ZK3- 
4709, quinazoline ZD6474, pyrrolocarbazole CEP-7055 (orally active N,Ndimethylglycine 
5 ester of CEP-5241), and CP-547632. VEGFR2 inhibitors are generally known or can be 
identified using VEGFR2 kinase assays, 

A. Second Agents 

Sorafenib can be combined with other phaxnciacologically active compounds ("second 
agents") in naethods and compositions of the invention. In a preferred embodiment, the 

10 second agents are capable of reducing pulmonary artery pressure or vascular resistance, 
inhibiting thrombosis or thromboembohsm, or ensuring compliance of patients. Examples of 
the second agents include, but are not limited to, anticoagulants, diuretics^, cardiac glycosides, 
calcium channel blockers, vasodilators, prostacyclin and prostacyclin analogues, endothelin 
receptor antagonists, phosphodiesterase inhibitors {e.g.^, PDE 5 inhibitors), endopeptidase 

15 inhibitors, lipid lowering agents, thromboxane inhibitors, and other therapeutics known to 
reduce pulmonary artery pressure. 

Specific second agents are anticoagulants, which are useful in the treatment of 
patients with PH who have an increased risk of thrombosis and thromboembolism. A 
particular anticoagulant is warfarin (Coumadin®). 

20 Other second agents include diuretics, cardiac glycosides, and oxygen. Digoxin 

therapy is used to improve right ventricular function in patients with right ventricular failure. 
Diuretics can be used to manage peripheral edema. Oxygen supplementation may be used in 
those patients with resting or exercise-induced hypoxemia. 

Calcium channel blockers such as diltiazem, amlodipine, and nifedipine can also be 
25 used as second agents, particularly for vasoreactive patients at right heart catheterization. 
These drugs are thought to act on the vascular smooth muscle to dilate the puhnonaiy 
resistance vessels and lower the pulmonary artery pressure (Tapson, 2002). 

Other second agents include vasodilators, particularly for NYHA/WHO class III and 
rv patients with right heart failure who do not respond to calcixim channel blockers or are 
30 unable to tolerate them. Examples of vasodilators include, but are not limited to, prostacyclin 

18 



wo 2007/087575 



PCT/US2007/060995 



(e.g., prostaglandin I2 (PGI2), epoprostenol (EPO, Flolan®), treprostinil (RemodulinCE))), and 
nitric oxide (NO). 

Still other second agents are endothelin antagonists. One example is bosentan 
(Tracleer®), which competitively binds to endothelin-1 (ET-1) receptors A + B, causing 
5 reduction in pulmonary artery pressure. 

Specific second agents used in the invention include;, but are not limited to, 
amlodipine, nifedipine, diltiazem, bosentaa (Tracleer®), prostacyclin {e,g., epoprostenol 
(Flolan®), treprostinil (Remodulin®), iloprost), warfarin (Coumadin®), tadalafil (Cialis®), 
simvastatin (Zocor®), omapatrilat (Vanlev®), irbesartan (Avapro®), pravastatin 
10 (Pravachol®), digoxin, nitric oxide, L-arginine, iloprost, betaprost, and sildenafil (Viagra®). 

IV. METHODS OF TREATMENT AND MANAGEMENT 

A. Diagnosis 

Pulmonary Hypertension is typically defined as a pulmonary artery mean pressure 
greater than 20 mm Hg with a pixlmonary vascular resistance greater than two Wood units. 

15 Pulmonary hypertension is indicated by increased shortness of breath during exertion 
accompanied by one of the known causes of PH. Methods of diagnosing PH include 
echocardiography, Doppler flow studies, assessment of blood oxygenation, pulomonary 
function, computer tomography of the chest, ventilation-perfusion lung scanning, and cardiac 
catheterization (Nauser and Stites, 2001), Pulmonary hypertension left untreated results in 

20 right ventricular failure and death. Diagnosis of PH may be associated with identification of 
right ventricular hypertrophy on an ECG or prominent pulmonary arteries on a chest 
radiograph. Once an indication of PH has been establish a patient will typically undergo two- 
dimensional echocardiography with Doppler flow studies. Typically:, PH is confirmed by 
identification of tricuspid regurgitation with right ventricular enlargement and/or dysfunction. 

25 Once a patient has been diagnosed with PH, the patient should undergo testing to 

identify any underlying causes for PH. The test include^ but are not limited to blood analysis, 
including blood count, prothrombin time, partial thromboplastin time, hepatic profile, 
autoimmune panel, basic naturetic peptide (BNP)^ and HIV testing; blood gas analysis; 
pulmonary ftmction testing; CT scan; ventilation-perfiasion lung scan; cardiac catheterization; 

19 



wo 2007/087575 



PCT/US2007/060995 



or combinations thereof (see Nauser and Stites, 2001 figure 3 algoritlun for evaluation of a 
patient with suspected pubnonary hypertension). 

The familial and medical history of a patient may be used to identify a subject at risk 
of developing PH and is a candidate for prophylactic or preventative treatments. 

5 B, Treatment 

Treatment of PH depends on the stage and the mechanism of the disease. Typical 
treatments for PH include, but are not limited to correction of underlj/ing cause (e.g.^ surgical 
treatment of mitral stenosis^ left to right shunt, or accessible chronic thromboemboli; 
afterload reduction, digoxin and diuretics for left ventricular dysfunction; prevention and 

10 treatment of respiratory infection; avoidance of anorectic agents); decrease pulmonary 
vasular resistance (e,g,^ vasodilation (oxygen, calcium channel blockers, prostacyclins, nitric 
oxide), or anticoagulation); increase cardiac output (short-term parenteral inotropes or 
digoxin); reduce volume overload (low-salt diet or diuretics); or surgery (lung transplant or 
atrial septosotomy) (see Nauser and Stites, 2001 table 5 possible treatments for pulmonary 

15 hypertension). 

Several studies suggest that survival is increased when the patient is treated with 
anticoagulant therapy, regardless of histopathologic subtype of PAH (Rubin et al, 1997). 
Warfarin is used to maintain an International Normalized Ratio of 1.5- to 2-times the control 
value, provided no contraindication to anticoagulation is present (Tapson, 2002). Warfarin is 
20 the standard of care for thromboembolic PH. 

Digoxin is used to prevent and treat supraventricular arrhythmias associated with 
PAH and for patients who have concomitant left heart failure. However, no randomized 
controlled clinical study has been performed to validate this strategy for patients with IP AH 
(Tapson, 2002). Diuretics are reportedly useful in reducing excessive preload in patients with 
25 right heart failure (Rubin et al, 1997). Oxygen supplementation is nsed in those patients 
with resting or exercise-induced hypoxemia (Rubin et al., 1997; Tap son, 2002). 

Arterial septostomy or lung transplant is indicated for patients who do not respond to 
medical therapy (The Merck Manual 1999; Rubin, 2002). Arterial septostomy is intended to 
serve as a bridge to transplantation. 



20 



wo 2007/087575 



PCT/US2007/060995 



Medications presently used for the treatment of PH include calcium channel blockers 
and pulmonary vasodilators (The Merck Manual 1999; Tapson, 2002). Calcium channel 
blockers are utilized for "true responders" as evidenced by response during right heart 
catheterization (McLaughlin et al^ 2004, which is incorporated herein by reference in its 
entirety). Vasodilators include, but are not limited to epoprostenol, treprostinil, and iloprost. 
Endothelin antagonists include, but are not limited to Tracleer and the like. 
Phosphodiesterase inhibitors include, but are not limited to sildanefil and the like. 

Metliods of this invention encompass methods of preventing, treating and/or 
managing various types of PH, particularly PAH. As used herein, imless otherwise specified, 
the term "preventing" or "prophylaxis" includes, but is not limited to, inhibiting or averting 
one or more symptoms associated with PH. Symptoms associated with PH include, but are 
not limited to, dyspnea, fatigue, weakness, chest pain, recurrent syncope, seizures, light- 
headedness, leg edema, and palpitations. As used herein, unless otherwise specified, the term 
"treating" refers to the administration of a composition after the onset of symptoms of PH, 
whereas "preventing" refers to the administration prior to the onset of symptoms, particularly 
to patients at risk of PH. As used herein and unless otherwise indicated, the term "managing" 
encompasses preventing the recurrence of PH in a patient who had suffered from PH, and/or 
lengthening the time that a patient who had suffered from PH remains in remission. 

The invention encompasses methods of treating or managing patients who have been 
previously treated for PH, as well as those who have not previously been treated for PH. 
Because patients with PH have heterogenous clinical manifestations and varying cUnical 
outcomes, it is preferred that patients should be treated according to the severity and stage of 
the disease. Methods and compositions of this invention can be used in various stages or 
types of PH including, but not limited to, primary PH, secondary PH and WHO classes I to 
IV patients. 

Methods encompassed by this invention comprise administering sorafenib, or a 
pharmaceutically acceptable salt or prodrug thereof to a patient {e.g., a human) suffering, or 
likely to suffer, firom PH. La one aspect of the invention, sorafenib is administered in single 
or divided daily doses in an amoimt of from about 0.1 to about 1000 mg/day, from about 100 
to about 800 mg/day, or from about 50 to about 400 mg/day. 

C. Combination Therapy 



21 



wo 2007/087575 



PCT/US2007/060995 



Administration of sorafenib and a second agent to a patient can occur simultaneously 
or sequentially by tlie same or different routes of administration. The suitability of a 
particular route of administration employed for a particular agent will depend on the agent 
itself (e,g.^ whether it can be administered orally without decomposing prior to entering the 
5 blood stream) and the disease being treated. A preferred route of administration for sorafenib 
is oraL Another preferred route of administration for sorafenib is parenteral^ particularly for 
patients who are in a peri-transplant period or in an end stage of PH. Preferred routes of 
administration for the second agent of the invention are known to those of ordinary skill in 
the art such as in Physicians' Desk Reference (2003). 

10 Various combinations may be employed^, sorafenib is "A" and a second agent is "B": 

A/B/A B/A/B B/B/A A/A/B A/B/B B/A/A A/B/B/B B/A/B/B 

B/B/B/A B/B/A/B A/A/B/B A/B/A/B A/B/B/A B/B/A/A 

B/A/B/A B/A/A/B A/A/A/B B/A/A/A A/B/A/A A/A/B/A 

Administration of the compositions of the present invention to a patient will follow 
1 5 general protocols for the administration of agents for the treatment of PAH. It is expected 
that the treatment cycles would be repeated as necessary. It also is contemplated that various 
standard therapies, as well as surgical intervention, may be applied in combination with the 
described compositions and formulations. 

The specific amount of the second agent will depend on the specific agent used, the 
20 type of PH being treated or managed, the severity and stage of PH, and the amount(s) of 
sorafenib and any optional additional agents concurrently administered to the patient. In 
specific embodiments of the invention, the second agent is amlodipine, diltiazem, nifedipine, 
prostacyclins (e.g-., epoprostenol (Flolan®), treprostinil (Remodulin®), iloprost), bosentan 
(Tracleer®), warfarin (Coumadin®), tadalafil (Cialis®), simvastatin (Zocor®), omapatrilat 
25 (Vanlev®), irbesartan (Avapro®), pravastatin (Pravachol®), digoxin, nitric oxide, L-arginine, 
beraprost, or sildenafil (Viagra®). 

In one embodiment of the invention, sorafenib is administered to reduce a period of 
treatment with a second agent typically used to treat PH. In a particular embodiment, at the 
beginning of week one, firom about 100 to about 800 mg/day of sorafenib is administered 



22 



wo 2007/087575 



PCT/US2007/060995 



along with a second agent in an amount that those of ordinary skill in the art can deteimine by 
their professional judgment- At the beginning of weeks 5, 9, 13, and 17^ withdrawal of the 
second agent may occur in increments of 25% of the initial dose of the second agent. At the 
beginning of week 17, dose of the second agent may be 800 mg/day if symptoms of a patient 
5 do not worsen. If symptoms of a patient worsen, dose of the second agent may be increased 
to stabilize the patient. 

D. Combination with Surgery or Transplantation 

This invention encompasses a method of treating or managing PH, which comprises 
administering sorafenib, or a pharmaceutically acceptable salt or prodmg thereof, in 
10 conjimction with surgery or transplantation therapy. As discussed herein, the treatment of PH 
varies, depending on the stage and mechanism of the disease. Arterial septostomy or lung 
transplantation may be necessary for PH patients who have failed to respond to other therapy. 
Sorafenib may provide additive or synergistic effects when given before, concurrently with, 
or after surgery or transplantation therapy in patients with PH. 

15 V. PHARMACEUTICALS AND METHODS FOR THE TREATMENT OF 

DISEASE 

hi additional embodiments, the present invention concerns formulation of sorafenib 
compositions disclosed herein in pharmaceutically-acceptable solutions for administration to 
a cell, tissue, animal, or patient either alone, or in combination with one or more second agent 
20 or second therapy. 

Aqueous pharmaceutical compositions of the present invention will have an effective 
amount of a sorafenib that modulates PH and/or its related pathologies or etiologies. Such 
compositions generally will be dissolved or dispersed in a pharmaceutically acceptable 
carrier or aqueous medium. An "effective amount,'^ for the purposes of therapy, is dejSned at 
25 that amount that causes a clinically measurable difference in the condition of the subject. 
This amount will vary depending on the substance, the condition of the patient, the type of 
treatment, etc. 

The phrases "pharmaceutically" or "pharmacologically acceptable" refer to molecular 
entities and compositions that do not produce a significant adverse, allergic or other untoward 
30 reaction when administered to an aaimal, or human, as appropriate. As used herein. 



23 



wo 2007/087575 



PCT/US2007/060995 



"pharmaceutically acceptable carrier" includes aiay aaid all solvents, dispersion media^ 
coatings, antibacterial and antifimgal agents, isotonic and absorption delaying agents and the 
like. The use of such media and agents for pharmaceutically active substances is well known 
in the art. Except insofar as any conventional media or agent is incompatible with the active 
5 ingredients, its use in the therapeutic compositions is contemplated. Supplementary active 
ingredients^ such as other anti-diabetic agents, can also be incorporated into the compositions. 

In addition to the compounds formulated for parenteral administration, such as those 
for intravenous or intramuscular injection^ other pharmaceutically acceptable forms include^ 
e.g., tablets or other solids for oral administration; time release capsules; and any other form 
1 0 currently used, including creams^ lotions, inhalants and the like. 

The active compounds of the present invention will often be formulated for parenteral 
administration, formulated for injection via the intravenous, intramuscular, 

subcutaneous, or even intraperitoneal routes. The preparation of a composition that contains 
sorafenib alone or in combination with a second therapeutic agent as active ingredients will 
15 be known to those of skill in the art in light of the present disclosure. Typically, such 
compositions can be prepared as injectables^ either as liquid solutions or suspensions; solid 
forms suitable for using to prepare solutions or suspensions upon the addition of a liquid prior 
to injection can also be prepared; and the preparations can also be emulsified. 

Solutions of the active compounds as free base or pharmacologically acceptable salts 
20 can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. 
Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures 
thereof and in oils. Under ordinary conditions of storage and use, these preparations contain 
a preservative to prevent the growth of microorganisms. 

The pharmaceutical forms suitable for injectable use include sterile aqueous solutions 
25 or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; 
and sterile powders for the extemporaneous preparation of sterile injectable solutions or 
dispersions, hi many cases, the form must be sterile and must be fluid to the extent that easy 
syringability exists. It must be stable under the conditions of manufacture and storage and 
must be preserved against the contaminating action of microorganisms, such as bacteria and 
30 fungi. 



24 



wo 2007/087575 



PCT/US2007/060995 



The active compounds may be fomiulated into a composition in a neutral or salt form. 
Pharmaceutically acceptable salts include the acid addition salts and which are formed with 
inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids 
as acetic, oxalic, tartaric, mandelic, and the like. Salts formed can also be derived from 
5 inorganic bases such as, for example, sodium, potassium, ammonium, calcium^ or ferric 
hydroxideS;, and such organic bases as isopropylamine, trimethylamine, histidine, procaine 
and the like. 

The carrier also can be a solvent or dispersion medium containing, for example, 
water, ethanolj polyol (for example, glycerol, propylene glycol, and liquid polyethylene 

10 glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be 
maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the 
required particle size in the case of dispersion and by the use of surfactants. The prevention 
of the action of microorganisms can be brought about by various antibacterial and antifungal 
agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In 

15 many cases, it will be preferable to include isotonic agents, for example, sugars or sodium 
chloride. Prolonged absorption of the injectable compositions can be hrought about by the 
use in the compositions of agents delaying absorption, for example, alrmiinum monostearate 
and gelatin. 

Sterile injectable solutions are prepared by incorporating the active compoimds in the 
20 required amount in the appropriate solvent with various other ingredients enumerated above, 
as required, followed by filtered sterilization. Generally, dispersions are prepared by 
incorporating the various sterilized active ingredients into a sterile vehicle which contains the 
basic dispersion medium and the required other ingredients from those enumerated above. In 
the case of sterile powders for the preparation of sterile injectable solutions, the preferred 
25 methods of preparation are vacuum-drying and fireeze-drying techniques which yield a 
powder of the active ingredient plus any additional desired ingredient from a previously 
sterile-filtered solution thereof. 

Upon formulation, solutions will be administered in a manner compatible with the 
dosage fomiulation and in such amount as is therapeutically effective. The formulations are 
30 easily administered in a variety of dosage forms, such as the type of injectable solutions 
described above, with even drug release capsules and the like being employable. 

25 



wo 2007/087575 



PCT/US2007/060995 



For parenteral administration in an aqueous solution, for example, the solution should 
be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient 
saline or glucose. These particular aqueous solutions are especially suitable for intravenous, 
intramusculaiv subcutaneous and intraperitoneal administration. In this connection, sterile 
aqueous media which can, be employed will be known to those of skill in the art in light of the 
present disclosure. For example, one dosage could be dissolved in 1 mL of isotonic NaCl 
solution and either added to 1000 mL of hypodermoclysis fluid or injected at the proposed 
site of infusion, (see for example, "Remington's Pharmaceutical Sciences" 1035-1038 and 
1570-1580). Some variation in dosage will necessarily occur depending on the condition of 
the subject being treated. The person responsible for administration will, in any events 
determine the appropriate dose for the individual subject. 

In certain aspects of the methods of the invention, the route that the therapeutic 
composition is administered may be by parenteral administration. The parenteral 
administration maybe intravenous injection, subcutaneous injection, intramuscular injection, 
ingestion or a combination thereof In certain aspects, the composition comprising sorafenib 
is administered from about 0,1 to about 10 micro gram/kg/body weight per dose. In certain 
aspects, the composition comprising sorafenib is administered from about 1 to about 5 
microgram/kg/body weight per dose. In certain aspects, the composition comprising 
sorafenib is administered from about 1.2 to about 2.4 microgram/kg/body weight per dose. In 
certain aspects, the amount of sorafenib administered per dose may be about 0.1, about 0.2, 
about 0.3, about 0.4, about 0.5, about 0,6, about 0.7, about 0,8, about 0.9, about LO, about 

1.1, about 1.2, about 1.3, about 1. 4, about 1.5, about L6, about 1.7, about 1.8, about 1.9, 
about 2.0, about 2,1, about 2.2, about 2.3, about 2.4, about 2.5, about 2.6, about 2. 7, about 

2.8, about 2.9. about 3.0, about 3.1, about 3.2, about 3.3, about 3.4, about 3 5, about 3.6, 
about 3.7, about 3.8, about 3.9, about 4,0, about 4.1, about 4.2, about 4.3, about 4.4, about 

4.5, about 4,6, about 4. 7, about 4.8, about 4.9, about 5.0, about 5.1, about 5.2, about 5.3, 
about 5.4, about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, about 6. 0, about 6.1, about 

6.2, about 6.3, about 6.4, about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, 
about 7.1, about 7.2, about 7. 3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 

7.9, about 8.0, about 8.1, about 8.2, about 8,3, about 8.4, about 8.5, about 8. 6, about 8.7, 
about 8.8, about 8.9, about 9.0, about 9.1, about 9.2, about 9.3, about 9.4, about 9.5, about 

9.6, about 9.7, about 9.8. about 9.9 to about 10.0 or more nanogram/kg/body, 
microgram/kg/body, or milligram/kg/body. 



26 



wo 2007/087575 PCT/US2007/060995 



Formulation of phannaceutically-acceptable excipients and carrier solutions is well- 
Icnown to those of skill in the art, as is the development of suitable dosing and treatment 
regimens for using the particular compositions described herein in a variety of treatment 
regimens^ including e.g.^ oral^ parenteral, intravenous, intranasal, and intramuscular 
5 administration and formulation. 

A- Alimentary Delivery 

The tenn "alimentary delivery" refers to the administration, directly or otherwise, to a 
portion of the alimentary canal of a subject or patient. The term "alimentary canal" refers to 
the tubular passage that functions in the digestion and absorption of food and the elimination 
of food residue, which runs from the mouth to the anus, and any and all of its portions or 
segment Ss e,g.^ the oral cavity, the esophagus, the stomach, the small and large intestines and 
the colon, as well as compound portions thereof such as, e.g., the gastro-intestinal tract. 
Thus, the term "alimentary delivery" encompasses several routes of administration including, 
but not limited to, oral, rectal, endoscopic and sublingual/buccal administration. A common 
requirement for these modes of administration is absorption over some portion or all of the 
alimentary tract and a need for efficient mucosal penetration of the agent so administered. 

1, Oral Delivery 

In certain applications, the pharmaceutical compositions disclosed herein may be 
delivered via oral administration to an animal. As such, these compositions may be 
20 formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed 
in hard- or soft- shell gelatin capsule, or they may be compressed into tablets, or they may be 
incorporated directly with the food of the diet. 

The active compoimds may even be incorporated with excipients and used in the form 
of ingestible tablets, buccal tables, troches, capsules, elixirs, suspensions, syrups, wafers, and 

25 the like (Mathiowitz et al, 1997; Hwang et ah, 1998; U.S. Patents 5,641,515; 5,580,579 and 
5,792,451, each specifically incorporated herein by reference in its entirety). The tablets, 
troches, pills, capsules and the like may also contain the following: a binder, as gum 
tragacanth, acacia, cornstarch, or gelatin; excipients, such as dicalcium phosphate; a 
disintegrating agent, such as com starch, potato starch, alginic acid and the like; a lubricant, 

30 such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin 
may be added or a flavoring agent, such as peppermint, oil of wintergreen, or cherry 

27 



10 



15 



wo 2007/087575 



PCT/US2007/060995 



flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of 
the above type, a liquid carrier. Various other materials may be present as coatings or to 
otherwise modify the physical form of the dosage imit. For instance, tablets, pills^ or 
capsules may be coated with shellac^ sugar, or both. A syrup or elixir may contain the active 
5 compound sucrose as a sweetening agent methyl and propylparabens as preservatives, a dye 
and flavoring, such as cherry or orange flavor. Of course, any material used in preparing any 
dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts 
employed. In addition, the active compoimds may be incorporated into sustained-release 
preparation and formulations. 

10 Typically, these formulations may contain at least about 0.1% of the active compound 

or more, although the percentage of the active ingredient(s) may, of course, be varied and 
may conveniently be between about 1 or 2% and about 60% or 70% or more of the weight or 
volume of the total formulation. Naturally, the amormt of active compound(s) in each 
therapeutically useful composition may be prepared is such a way that a suitable dosage will 

15 be obtained in any given unit dose of the compound. Factors such as solubility, 
bioavailability-, biological half-life, route of administration, product shelf life, as well as other 
pharmacological considerations will be contemplated by one skilled in the art of preparing 
such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens 
may be desirable. 

20 2. Endoscopic Administration 

Endoscopy can be used for therapeutic delivery directly to an interior portion of the 
alimentary tract. For example, endoscopic retrograde cystopancreatography (ERCP) takes 
advantage of extended gastroscopy and permits selective access to the biliary tract and the 
pancreatic duct (Hirahata et al.^ 1992). However, the procedure is unpleasant for the patient, 
25 and requires a highly skilled staff. 

3. Rectal Administration 

Therapeutics administered by the oral route can often be alternatively administered by 
the lower enteral route, t e. , through the anal portal into the rectum or lower intestine. Rectal 
suppositories, retention enemas or rectal catheters can be used for this pmpose and may be 
30 preferred when patient compliance might a otherwise be difficult to achieve {e.g,^ in pediatric 
and geriatric applications, or when the patient is vomiting or unconscious). Rectal 

28 



wo 2007/087575 



PCT/US2007/060995 



administration may result in more prompt and higher blood levels than the oral route, but the 
converse may be true as well (Remington's Pharmaceutical Sciences, 711, 1990). Because 
about 50% of the tlierapeutic that is absorbed from the rectum will bypass the liver, 
administration by this route significantly reduces the potential for first-pass metabolism 
5 (Ben&tet al., 1996). 

B. In j ectable Delivery 

In certain circumstances it will be desirable to deliver the pharmaceutical 
compositions disclosed herein parenterally, intravenously, intramuscularly, or even 
intraperitoneally as described in U.S. Patents 5,543,158; 5,641,515 and 5,399,363 (each 

10 specifically incorporated herein by reference in its entirety). Solutions of the active 
compounds as free base or pharmacologically acceptable salts may be prepared in water 
suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be 
prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under 
ordinary conditions of storage and use, these preparations contain a preservative to prevent 

15 the growth of microorganisms. 

The pharmaceutical forms suitable for injectable use include sterile aqueous solutions 
or dispersions and sterile powders for the extemporaneous preparation of sterile injectable 
solutions or dispersions (U.S. Patent 5,466,468, specifically incorporated herein by reference 
in its entirety). In all cases the form must be sterile and must be fluid to the extent that it is 
20 easy to use a syringe. 

For parenteral administration in an aqueous solution, for example, the solution should 
be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient 
saline or glucose. These particular aqueous solutions are especially suitable for intravenous, 
intramuscular, subcutaneous and intraperitoneal administration (see for example. Remington's 

25 Pharmaceixtical Sciences, 1035-1038 and 1570-1580. Some variation in dosage will 
necessarily occur depending on the condition of the subject being treated. The person 
responsible for administration will, in any event, determine the appropriate dose for the 
individual subject. Moreover, for himian administration, preparations should meet sterility, 
pyrogenicity, and the general safety and purity standards as required by FDA Office of 

30 Biologies standards. 



29 



wo 2007/087575 



PCT/US2007/060995 



4. Parenteral Delivery 

The term "parenteral delivery" refers to the administration of a therapeutic of the 
invention to an animal in a manner other than through the digestive canal. Means of 
preparing and administering parenteral pharmaceutical compositions are loiown in the art 
5 (see, e.g.. Remington's Pharmaceutical Sciences, pages 1545-1569, 1990). 

5. Intraluminal administration 

Intraluminal administration, for the direct delivery of a therapeutic to an isolated 
portion of a tubular organ or tissue {e,g.y such as an artery, vein, ureter or urethra), may be 
desired for the treatment of patients with diseases or conditions afflicting the lirnien of such 

1 0 organs or tissues. To effect this mode of administration, a catheter or cannula is surgically 
introduced by appropriate means. After isolation of a portion of the tubular organ or tissue 
for which treatment is sought, a composition comprising a therapeutic of the invention is 
infused through the cannula or catheter into the isolated segment. After incubation for fi-om 
about 1 to about 120 minutes, during which the therapeutic is taken up or in contact with the 

15 cells of the interior lumen of the vessel, the infusion cannula or catheter is removed and flow 
within the tubular organ or tissue is restored by removal of the ligatures which effected the 
isolation of a segment thereof (Morishita et al^ 1993). Therapeutic compositions of the 
invention may also be combined with a biocompatible matrix, such as a hydrogel material, 
and applied directly to vascular tissue in vivo. 

20 C. Nasal Delivery 

In certain embodiments, the pharmaceutical compositions may be delivered by 
intranasal sprays, inhalation, and/or other aerosol delivery vehicles. Methods for delivering 
genes, nucleic acids, and peptide compositions directly to the lungs via nasal aerosol sprays 
has been described e.g.^ in U.S. Patents 5,756,353 and 5,804,212 (each specifically 

25 incorporated herein by reference in its entirety). Likewise, the delivery of dmgs using 
intranasal microparticle resins (Takenaga et ah, 1998) and lysophosphatidyl-glycerol 
compounds (U.S, Patent 5,725,871, specifically incorporated herein by reference in its 
entirety) axe also well-known in the pharmaceutical arts. Likewise, transmucosal drug 
delivery in the form of a polytetrafluoroetheylene support matrix is described in U.S. Patent 

30 5,780,045 (specifically incorporated herein by reference in its entirety). 

30 



wo 2007/087575 



PCT/US2007/060995 



D. Epidermal and Transdermal Delivery 

Epidermal and Transdermal Delivery, in which pharmaceutical compositions 
containing therapeutics are applied topically, can be used to administer drugs to be absorbed 
by the local dermis or for further penetration and absorption by underlying tissues, 
5 respectively. Means of preparing and administering medications topically are known in the 
art (see^ e.g.^ Remington's Pharmaceutical Sciences, 1596-1609;, 1990). 

E. Liposome-, Nanocapsule-, and Microparticle-Mediated Delivery 

In certain embodiments, the inventors contemplate the use of liposomes, 
nanocapsules, microparticles, microspheres, lipid particles, vesicles, and the like, for the 
10 introduction of the compositions of the present invention into suitable host cells. la 
particular, the compositions of the present invention may be formulated for delivery either 
encapsulated in a lipid particle, a liposome, a vesicle^ a nanosphere, or a nanoparticle or the 
like. 

Such formulations may be preferred for the introduction of pharmaceutically- 
15 acceptable formulations of the nucleic acids or constmcts disclosed herein. The formation 
and use of liposomes is generally known to those of skill in the art (see for example, 
Couvreur et aL, 1977; Couvreur, 1988; Lasic, 1998; which describes the use of liposomes and 
nanocapsules in the targeted antibiotic therapy for intracellular bacterial infections and 
diseases). Recently^ liposomes were developed with improved serum stability and circulation 
20 half-times (Gabizon and Papahadjoponlos, 1988; Allen and Chorua, 1987; U.S, Patent 
5,741^516, specifically incorporated herein by reference in its entirety). Further^ various 
methods of liposome and liposome like preparations as potential drug carriers have been 
reviewed (Takakura, 1998; Chandran et ah, 1997; Margalit, 1995; U.S. Patent 5,567,434; 
5,552,157; 5,565,213; 5,738,868 and 5, 795,587, each specifically incorporated herein by 
25 reference in its entirety). 

Liposomes are formed firom phospholipids that are dispersed in an aqueous medium 
and spontaneously form multilamellar concentric bilayer vesicles (also termed multilamellar 
vesicles (MLVs). MLVs generally have diameters of firom 25 nm to 4 |Lim. Sonication of 
MLVs results in the formation of small imilamellar vesicles (SUVs) with diameters in the 
30 range of 200 to 500 A, containing an aqueous solution in the core. 



31 



wo 2007/087575 



PCT/US2007/060995 



The fate and disposition of intravenously injected liposomes depend on their physical 
properties, such as size, fluidity, and surface charge. They may persist in tissues for h or 
days, depending on their composition, and half Uves in the blood range firom min to several h. 
Larger liposomes, such as MLVs and LUVs, are taken up rapidly by phagocytic cells of the 
5 reticuloendothelial system, but physiology of the circulatory system restrains the exit of such 
large species at most sites. They can exit only in places where large openings or pores exist 
in the capillary endothelium, such as the sinusoids of the liver or spleen. Thus, these organs 
are the predominate site of uptake. On the other hand^ SUVs show a broader tissue 
distribution but still are sequestered highly in the liver and spleen. In general, this in vivo 
10 behavior limits the potential targeting of liposomes to only those organs and tissues 
accessible to their large size. These include the blood, liver, spleen, bone marrow, and 
lymphoid organs. 

Alternatively, the invention provides for pharmaceutically-acceptable nanocapsule 
formulations of the compositions of the present invention. Nanocapsules can generally 

15 entrap compounds in a stable and reproducible way (Henry-Michelland et aL^ 1987; 
Quintanar-Guerrero et al.^ 1998; Douglas et aL, 1987). To avoid side effects due to 
intracellular polymeric overloading, such ultrafine particles (sized around 0.1 [mgr]m) should 
be designed using polymers able to be degraded in vivo. Biodegradable polyalkyl- 
cyanoacrylate nanoparticles that meet these requirements are contemplated for use in the 

20 present invention. Such particles may be are easily made, as described (Couvreur et ah^ 1980; 
1988; zur Muhlen et aL, 1998; Zambaux et al, 1998; Pinto-Alphandary et al. , 1995 and U.S. 
Patent 5,145,684, specifically incorporated herein by reference in its entirety). 

VI. KITS 

In some cases, agents of the invention are not administered to a patient at the same 
25 time or by the same route of administration. This invention therefore encompasses kits 
which, when used by the medical practitioner, can simplify the administration of appropriate 
amounts of agents to a patient, 

A typical kit or composition of the invention comprises a dosage form of sorafenib, or 
a pharmaceutically acceptable salt or prodrug thereof. Klits encompassed by this invention 
30 can further comprise additional active agents such as amlodipine, dilitazem, nifedipine, 
adenosine, epoprostenol (Flolan®), treprostinil (Remodulin®), bosentan (Tracleer®), 

32 



wo 2007/087575 



PCT/US2007/060995 



warfarin (Coumadin®), tadalafil (Cialis®), simvastatin (Zocor®), omapatrilat (Vanlev®), 
irbesartan (Avapro®), pravastatin (Pravachol®), digoxin, nitric oxide, L-arginine, iloprost, 
beraprost, and sildenafil (Viagra®), or a combination thereof. Examples of the additional 
active agents include, but are not limited to, those disclosed herein. 

5 Kits of the invention can further comprise devices that are used to administer the 

active agents. Examples of such devices include, but are not limited to, syringes, drip bags, 
patches, and inhalers. 

Elits of the invention can farther comprise pharmaceutically acceptable vehicles that 
can be used to administer one or more active agents. For example, if an active agent is 

10 provided in a solid form that must be reconstituted for parenteral administration, the kit can 
comprise a sealed container of a suitable vehicle in which the active agent can be dissolved to 
form a particulate-free sterile solution that is suitable for parenteral administration. Examples 
of pharmaceutically acceptable vehicles include, but are not limited to: Water for Injection 
USP; aqueous vehicles such as, but not limited to. Sodium Chloride Injection, Ringer's 

15 Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer*s 
Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene 
glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, com 
oilj cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl 
benzoate. 

20 EXAMPLES 

The following examples are included to demonstrate embodiments of the invention. 
It should be appreciated by those of skill in the art that the techniques disclosed in the 
examples which follow represent techniques discovered by the inventor to function well in 
the practice of the invention, and thus can be considered to constitute preferred modes for its 
25 practice. However, those of skill in the art should, in light of the present disclosure, 
appreciate that many changes can be made in the specific embodiments which are disclosed 
and still obtain a like or similar result without departing from the spirit and scope of the 
invention. 



33 



wo 2007/087575 



PCT/US2007/060995 



EXAMPLE 1 

Pulmonary Hypertension Connection Database/Pulmonary Hypertension Specimen 

Collection 

The Pulmonary Hypertension Connection (PHC) has collected baseline demographic, 
5 clinical phenotype, medication^ echocardiography, exercise test, and cardiac catheterization 
data for 900 patients followed by the inventors practice since its inception in 1980- 
Development of this database began at Rush and has continued at the University of Chicago, 
Currently established at the University of Chicago^, the Pulmonary Hypertension Program has 
over 1200 active patients. Since receiving IRB-approval over 1,000 active patients have 
10 provided informed consent and a fiill-time data manager continues to input data on past and 
active patients. The PHC stores clinical data but also provides statistical analysis software in 
the single unit for rapid analysis by a trained investigator. 

In January 2005, the inventors' protocol to generate a longitudinal specimen 
collection entitled, "Biomarkers hi Pulmonary Hypertension" received IRB approval. The 

15 protocol is for collection of DNA, peripheral blood specimens, and clinically indicated 
central blood specimens. Discovery of relevant biomarkers in these specimens can be 
immediately and safely integrated into a prospective study and routine clinical care. Patients 
evaluated by pulmonary hypertension specialists who consent to the collection and archiving 
of plasma, serum, and DNA are currently enrolled. Blood is collected during clinically 

20 indicated evaluations either from a peripheral vein at scheduled office visits or from central 
catheters placed during cardiac catheterization. Plasma, serum, and DNA are isolated, frozen 
and stored by standard techniques in accord with International Society for Biological and 
Environmental Repositories (ISGER) best practices (Somiari et al.^ 2004; Friede et aL^ 2003). 
Coding of these specimens allows for retrospective selection of appropriate specimens to test 

25 potential biomarkers for prediction/association with specific clinical phenotypes identified in 
the PHC. 

EXAMPLE 2 

Development of Novel Endpoints 

The clinical assessment of patients with pulmonary hypertension currently involves an 
30 assessment of exercise capacity, echocardiography, and cardiac hemodynamics. Overall 
assessment is based on a combination of these factors but there have not been any guidelines 



34 



wo 2007/087575 



PCT/US2007/060995 



on treatment based on the overall assessment, only based on worsening functional class and 
cardiac hemodynamics. Invasive hemodynamics is still the most reliable and most accurate 
assessment of pulmonary vascular resistance. Catheterization does not provide information 
on the patient's functional ability, right ventricular or pulmonary vascular reserve^ or on 
5 pathologic vascular remodeling of the pulmonary circulation (regression, identification of 
plexiform lesions). Alternative non-invasive techniques may prove to be better biomarkers 
for PAH. The following experimental endpoints will be evaluated as novel measures of 
disease activity: Naughton-Balke treadmill test (TT), 3DE, and 64 slice CT scan. 

Naiighton-Balke Treadmill Test (TT) - 6MW is preformed by the subject walking 
10 on a flat surface at his/her own pace. The 6MW is not a true measure of exercise from a 
physiology perspective. Cardiopulmonary exercise testing in few centers is a better correlate 
of exercise physiology and hemodynamics, (Oudiz et al.^ 2006; Yasunobu et ah, 2005) but 
this test is time consuming, expensive, and difficult to do with reproducibiUty in 
unexperienced centers. The University of Chicago center is gaining expertise in this non- 
15 invasive test and may in the future incorporate this testing in protocols. The metabolic 
equivalent, or MET, is the amount of oxygen used by a seated person and is a predictor of 
survival in the general population. It is a physiologic measure of exercise capacity. The 
Naughton-Balke exercise protocol, (Patterson et al.^ 1972) measured in METs is easy to 
administer by programming a standard treadmill, is easily reproducible, inexpensive, and has 
20 been preformed by programs at the University of Chicago for many years. Because of the 
inventors familiarity with this test, and because it is a better measure of exercise physiology 
than the 6MW, it is contemplated that the MET will be a better predictor and measure to 
follow for PH patients. 

Using data from the Intravenous Treprostinil, study I determined the reliability of 
25 MET compared to the 6MW. Both tests were done by each patient in the study at multiple 
time points. Pearson correlation coefficient (r) was calculated to express inter-relationship 
between measures of exercise capacity. Intra-class correlation coefficient (ICC) was used to 
indicate the repeated reliability of exercise capacity measures over time. 6MW and MET 
were symmetrically distributed and correlated (r=0.63). ICCs for 6MW, TT, and MET were 
30 0.77, 0.82, and 0.80, respectively. Fimctional class correlated with 6MW and MET at 
baseline and at wk 12 (all p<0.01), but hemodynamics did not correlate with exercise 
measures consistently. Based on this result, MET is as reliable and reproducible as the 6MW. 

35 



wo 2007/087575 



PCT/US2007/060995 



The next step is to prove that MET is a better predictor aiid surrogate endpoint than the 
6MW. 

Three-Dimensional Echocardiography (3DE) - DE software (TomTec, Germany) 
has been developed that semi-automatically detects RV endocardial borders and addresses 
5 drawbacks of prior RV quantification. Patients have RT3DE as part of their routine 
evaluation with two-dimensional echocardiography (2DE). Investigation of the influence of 
different degrees of pulmonary hypertension on RV remodeling in patients and development 
of novel indices that may better assess this phenomenon are under way. Using a matrix array 
probe and Sonos 7500 (Philips, MA), a wide-angled acquisition of the RV during a breath 
10 hold gated to ECG will be obtained. The RV volume will be analyzed using software that 
semi-automatically detects RV endocardial borders resulting in global and regional RV 
volume and ejection fraction (EF). Several RV indices will include RV diameter in 2D and 
3D, 3D area measurements at the tricuspid valve (TV) annulus, 1/3 of the RV and 2/3 of the 
RV. 

15 In initial observations 3D echocardiographers at flie University of Chicago, found that 

RV remodeling is most evident at 1/3 of the RV. The TV annulus does not significantly 
dilate until patients have severe PH. The inventors anticipate that this technology will 
expand the knowledge of how the RV reacts towairds changes in pressure^ volume overload or 
different disease states. Patients will continue to have RT3DE in Phase I/II trials to assess 

20 and monitor response to therapy. 

Computed Tomography (CT) Scaus - At the University of Chicago under the 
direction of a leading radiologist in computer based analysis of radiographic imaging; the 
inventors have developed a fully automated lung segmentation method for thoracic CT scans 
that has been used for automated analysis of lung parenchyma texture, including the lung 

25 texture of patients with PH. A preliminary study of 28 CT scans (14 from PH patients and 14 
from aged-matched "normal" patients) was conducted. The lung texture analysis method was 
applied to a single section from each CT scan; these individual sections were manually 
selected at the level of the bifurcation of the pulmonary artery. The lung texture features 
were used to discriminate between the normal and the PH groups. This discrimination task 

30 was performed by a linear discriminant classifier^ the performance of which was evaluated by 
receiver operating characteristic (ROC) analysis with the LABROC4 program (C.E. Metz^ 
Ph.D, The University of Chicago). The automated method achieved an area under the ROC 



36 



wo 2007/087575 



PCT/US2007/060995 



curve of 0.97 (out of a maximum 1.0) in this discrimination task (submitted ERS). Based on 
this preliminary study^ all patients seen by our PH center are asked to enroll in the CT 
database-a repository allowing high level analyses of scans. Shortly all PH patients with 
clinical indications for a chest CT will have a 64 shoe CT to continue work on this project 
5 and to detennine the ability to detect plexifomi lesions. 

EXAMPLE 3 

Preclinical Data with Dahl-Sensitive Rats 

Human Pulmonary Hypertension (PH) is an often fatal vascular disorder affecting -2- 
3 new cases/million/ yr characterized by increased pulmonary artery pressure, right heart 

10 failure, and death. Current strategies to treat PH are problematic and do not address the 
vascular remodeling (smooth muscle contraction, hypertrophy) characteristic of the disease. 
PH exhibits signijBcant overlap with cancer pathophysiology with abnormalities in signal 
transduction and cellular proliferation (endothelium, smooth muscle) resulting in an 
angioproliferative vasculopathy. As cancer and pulmonary hypertension share the 

15 involvement of angiogenesis and growth factor pathways, it was hypothesized that sorafenib 
may represent a novel therapeutic agent for PAH. The inventors set out to conduct pre- 
clinical studies to evaluate the safety and efficacy of sorafenib in a rodent model of hypoxia- 
induced PAH. This rat model combines hypoxia and SU5416, a VEGFR-2 inhibitor, which 
together produce PH characterized by: vasoconstriction, elevated pulmonary artery pressure, 

20 right ventricular hypertrophy, and vascular remodeling. (Taraseviciene-Stewart et ah FASEB 
J. 2001;15;427-.438). Another feature of this model is smooth muscle medial thickening and 
endothelial cell apoptosis. 

Protocol includes an initial administration of SU5416 subcutaneously at 20 mg/kg 
followed daily by sorenfenib oral daily doses of 2.5 mg/kg. Some animals were exposed to 

25 chronic hypoxia (10% O2)- The animals were divided into five experimental groups: hypoxia 
"i- sorafenib; hypoxia + SU5416 + sorafenib^ normoxia + vehicle control; Hypoxia -t- vehicle; 
and hypoxia 4- SU5416. PAP, RVSP, Echo, and DNA microarrays were used to measure 
various physiological and molecular biological parameters. FIG. 4 illustrates that 
hypoxia/SU5416-induces increases in right heart pressures and right heart hypertrophy 

30 (RV/LV+Septum). FIG. 4 also illustrates that sorafenib prevented hypoxia s- SU5416 
development of right heart hypertrophy (RV/LV+Septum) in Dahl SS rats. FIG, IB 
illustrates that sorafenib prevents hypoxia + SU5416 induced pulmonary hypertension and 

37 



wo 2007/087575 



PCT/US2007/060995 



remodeling. Sorafenib Prevents Remodeling in Dahl SS with PH (Health Edwards Grading) 
H/E sections (FIG. 17). 

To study potential mechanisms of sorafenib effects on PH RNA was isolated from rat 
lung tissue and analyzed using the Rat Chip II (Affymetrix). Sorafenib attenuated 
5 physiologic and histopathologic changes in PAH in a rodent model of PH. Mechanisms of 
PAH pathobiology is poorly understood therefore^ to explore potential mechanistic pathways 
the inventors conducted bioinformatic studies and expression profiling. Expression profiling 
revealed at least 179 genes differentially regulated by sorafenib^ when comparing profiling 
data to normoxia using GCRMA normalization in R and SAM (6 > 0.639, minimum fold 

10 change > 1.7). The candidate genes identified fell into various gene ontology classes, for 
example 4% were in the lung development and growth factors class (e.g., Bambi, Tgfl33:, 
Ltbp2), 13% in the cell migration/ECM class (e.g., Itga3_predicted, Cspg4j Cxcll2, Reln^ 
CoUSal, Cthrcl^ Hmgcr^ Cnnl^ Col6a2, Jag2), 4% in the apoptosis class (e,g,, Anzal, 
CoUSal, Jag2), 4% in the smooth muscle/fiber formation class (e.g,^ Des^ TpmS, Jph2)5 10% 

15 in the cell proliferation class (e.g., Anxal, Timpl, Lamb_l predicted, Cspg4, Cxcll2, Gja4, 
Jag2, Tgfb3), 6% in the blood vessel development class (e.g,, 0x0112, Cspg4, Gja4, CollSal, 
Serpinel), and 5% in the angiogenesis class (Cxcll2, Cspg4, Coll Sal, Serpinel). 



Table 2 Gene ontology for sorafenib influenced gene, genes known to be involved in PH, 









PubMatrix Terms 


Gene 
Symbol 


Gene Name 


Pulmonarv 
Hypertension 


Vascular 
Remodeling 


Hypoxia 


Endothelium 


DBS 


desmin 


103 


33 


308 


585 


McptlO 


Mast cell protease 10 




3 


2 


13 


Timpl 


Tissue inhibitor of 
metalloprotease 1 


11 


71 


30 


118 


Fbnl 


Fibrillin 1 


2 


5 


0 


18 


Nuprl 


Nuclear protein 1 


104 


140 


1680 


1646 


Tgfb3 


Transforming growth 
factor beta 3 


42 


111 


102 


442 


Bmprla 


Bone morphogenetic 
protein receptor, type 
lA 


8 


2 


1 


4 


Bmpr2 


Bone morphogenetic 
protein receptor, type n 


121 


10 


7 


18 


Itga6 


Integrin, alpha 6 


2 


11 


17 


364 


Tipc6 


Transient receptor 
potential cation channel, 
subfamily C member 


5 


3 


3 


9 



38 



wo 2007/087575 PCT/US2007/060995 



Table 3 Gene ontology for sorafenib influenced genes, unknown genes 





UnkIlo^?m genes 


PubMatrix Terms 


Gene symbol 


Gene name 


Pulmonary 
hypertension 


Vascular 
remodeling 


Hypoxia 


Endothelium 


Ankrdl 


Anlcyrin repeat domain 1 


0 


0 


3 


4 


Cnnl 


Calponinl 


0 


7 


2 


22 


Chstl2 


Carbolivdrate 
sulfo transferase 12 


0 


0 


0 


1 


T-Ttt! p*cr 1 


3 -livdroxv-3 - 
methylglutaryl- 
coenzvme A synthase 1 


0 


0 


0 


1 




Cvtochrome P450, 
subfamily 5 1 


0 


0 


0 


2 


Esml 


EndotheHal cell-specific 
molecule 1 


0 


1 


4 


53 


Gja4 


Gap junction membrane 
channel protein alpha 4 


0 


0 


0 


1 


Cxcll2 


Chemokine (C-X-C 
motif) ligand 12 


0 


14 


26 


149 


Jam3 


Junctional adhesion 
molecule 3 


0 


0 


1 


14 


Anxal 


Annexin Al 


0 


1 


2 


27 


Ltbp2 


Latent transforming 
growth factor beta 
binding protein 2 


0 


2 


1 


12 


Mgll 


Macrophage galactose 
N-acetyi-galactosamine 
specific lectin 1 


0 


0 


0 


2 



The inventors have successfully utilized sorafenib to attenuate (physiologic and 
histopathologic) the development of rodent pulmonary hypertension model (Dahl SS) 
5 utilizing chronic hypoxia and SU5416 administration. Gene expression profiling studies 
identified genes which are well recognized to be involved in angiogenesis, endothelial cell 
apoptosis, and PH (Table 2), as well as genes previously not associated with PH representing 
potential novel candidate genes (Table 3). Given its safety in advanced cancer patients, 
further studies exploring both the mechanism of action as well as human studieS;, evaluating 
10 sorafenib safety and efficacy in PH should be pxirsued. 

Preclinical data with Dahl-sensitive rats given sorefenib with hypoxia and SU5416 
did not develop evidence of PAH. Sorafenib may have a beneficial effect in the treatment 
PAH, The PHC database enables the phenotype our patients to be determined based on 
clinical data, which will be a useful tool in discriminating response to therapy. Non-invasive 
15 methods that may better evaluate and prove to be better screening and biomarkers in PAH 
include exercise treadmill testing by a Naughton-Balke protocol, 3DE, and CT scanning. 



39 



wo 2007/087575 



PCT/US2007/060995 



EXAMPLE 4 
Clinical Studies 

The pathophysiology of PAH overlaps the pathophysiology of cancer with aberrancies 
in signal transduction leading to abnormal endothelial and smooth muscle cell interactions 
5 and to angioproliferative vasculopathy. New signal transduction inhibitors being evaluated 
for the treatment of cancer represent potential effective therapies for PAH. With an existing 
phase I cancer therapeutics program, the University of Chicago provides a iinique 
environment to improve PAH care. Having established that the recently FDA-approved 
agent, sorafenib, an inhibitor of multiple kinases important to angiogenesis (Raf-1 kinase 
10 inhibitor, VEGFR-2, VEGFR-3, PDGFR-g), protected rats from developing PAH in the 
SU5416/hypoxia model of PAH, the inventors contemplate evaluation of this agent in Phase I 
and Phase II trials, 

A. Single center Phase IB trial of sorafenib in PAH patients. 

Study Objectives Phase IB study: To determine the toler ability of oral daily sorafenib 
15 in combination with prostacyclin sildenafil in pulmonary hypertension patients. This 
design is a single-center^ Phase IB dose escalation study of sorafenib up to the known MTD 
of 400 mg twice daily. Administration will continue until the occurrence of unacceptable 
toxicity (described below)^ withdrawn consent, disease progression, hospitalization for 
PH/iight heart failure, lung transplantation, or death. 

20 Accrual: The PH program is currently following over 150 subjects on stable 

prostacyclin therapy. Phase I will enroll 12 subjects and be completed within 6-9 months. 

Rationale for Inclusion Criteria: Prostacyclin therapy is the most efjacacious therapy 
ciorrently available for PAH. Long-term prostacyclin replacement is supported by the 
pathophysiology of PAH, the relative lack of prostacyclin seen in PAH, and prostacyclin's 
25 positive effects on pulmonary vascular bed. However, prostacyclin therapy is not curative. 
As a class, the side effect profile of all prostacyclins is similar to that of epoprostenol and is 
usually minimal and well tolerated in most subjects. These include flushing, headache, 
nausea^ loose stool, jaw discomfort with "first bite", and foot pain with prolonged standing or 
walking (Barst et ah, 1996; McLaughlin et ah, 2002; Sitbon et ah, 2002). 



40 



wo 2007/087575 



PCT/US2007/060995 



Epoprostenol, the first approved tlierapy for PAH, has a half-life of no more than 1-2 
minutes, mandating continuous intravenous therapy via a central catheter (Data et al.^ 1981). 
Cohort analyses in Europe and in the United States have provided convincing evidence of its 
long term benefits (McLaughlin et al^ 2002; Sitbon et al^ 2002). Because of its 
phannacology, prostacyclin analogues were developed to ease administration. Treprostinil is 
a tricyclic benzidene prostacyclin analogue that shares pharmacologic actions with 
epoprostenol (Clapp et aL, 2002) Treprostinil differs firom epoprostenol in that it is 
chemically stable at room temperature and neutral pH and has a longer half-life (3-4 hours) 
(Wade et al., 2004), Subcutanous treprostinir was approved in 2002 for the treatment of New 
York Heart Association (NYHA) class II-IV PAH patients (Simonneau et al, 2002) and 
recently, the FDA approved the use of intravenous treprostinil based on bioequivalence to 
subcutaneous therapy (Laliberte et al, 2004). Iloprost is a chemically stable prostacyclin 
analog that can be delivered by inhaler/nebulizer (Hoeper et al, 2000). This allowed a 
targeted approach, with direct inhalation of prostacyclin for more selective pulmonary 
effects. But because of its short duration of action it must be inhaled 6-12 times daily 
(Olschewski et aL, 2002; Hoeper etaL, 2000). 

To enhance prostacyclin benefits, combination therapeutic approaches and inhibiting 
multiple pathways concurrently may produce additive benefit. Others have examined 
combining a prostacyclin with agents that increase cyclic guanasine phosphate (cGMP). This 
is accomplished by inhibition of the phosphodiesterase 5 enzyme (PDE-5) which degrades 
cGMP in the vascular smooth muscle cell. The pulmonary vasculature has a higher 
concentration of the PDE-5 enzyme than most vascular beds. Oral sildenafil in combination 
with iloprost in (Hoeper et al, 2000) PAH patients over 9 to 12 months follow-up improved 
exercise capacity and hemodynamics (Ghofrani et al^ 2003). Open uncontrolled experience 
adding sildenafil to epoprostenol also improved hemodynamics (Stiebellehner et al^ 2003) 
and open label addition to subcutaneous treprostinil improved exercise capacity (Gomberg- 
Maitland et aL^ 2005). Large multicenter trials are currently in progress. Of note, animal 
data obtained prior to the large scale epoprostenol plus sildenafiil study consisted of safety 
and efficacy of sildenafil not the dmgs in combination. There is a reported animal study with 
beraprost (oral formulation not approved in U.S) plus sildenafil using the monocrotaline rat 
model that did not demonstrate any safety/toxicity and demonstrated efficacy and improved 
survival (Itoh et al^ 2004). Previous sorafenib studies have not demonstrated known dmg 
interactions with warfarin or digoxin. Patients on other classes of PAH therapy (endothelin 



41 



wo 2007/087575 



PCT/US2007/060995 



receptor antagonists, m-ginine), or experimental therapies will not be included based on the 
lack of conclusive data and on the preference to examine the drug in a homogenous patient 
population. 

PAH is a devastating disease. Ethically, for Phase I/II trials based on current 
5 available therapeutic agents, the inventors believe that all subjects should have significant 
exercise capacity limitation based on the 6MW and be receiving prostacyclin therapy with or 
without sildenafil. 

Patient Selection-Eligibility Criteria: 

1 . Age >1 8 years 

10 2, PAH as defined as IP AH, FPAH or PAH associated with connective tissue disease. 

(Humbert et al^ 2004) 

3. Baseline 6MW > 150 meters and < 450 meters 

4. PAH as defined by hemodynamics at diagnosis by right heart catheterization 
defined as: mean PAP >25 mmHg with a normal PCWP <15 mm Hg at rest and a PVR >2 

15 Woodimits 

5. Receiving conventional therapy as clinically indicated (oxygen, diuretics, 
aldosterone antagonist, calcium channel blockers, digoxin) with dose that is unchanged in the 
preceding 30 days prior to enrollment. This is excluding anticoagulants (warfarin) as the 
patient^s dose may not be stable if the patient is having a cardiac catheterization at baseline 

20 within 30 days of enrollment and warfarin is being held. The dose of warfarin needs to be 
stable for 7 days or therapeutic with an rNR=2.0 

6. On intravenous/subcutaneous prostacyclin at a stable dose > 30 days 

7. Subjects must be on sildenafil at a stable dose >30 days, 

8. Must have right heart catheterization on prostacyclin + sildenafil within preceding 
25 30 days. Subjects must be on a stable dose of medication within 30 days prior to cardiac 

catheterization and therefore there can be no dosage changes of the medications between 
catheterization and baseline. 

42 



wo 2007/087575 



PCT/US2007/060995 



9. Must have pulmonary function tests (PFT) within 90 days prior to enrolhnent: 
TLC, FEVl, FVC, DLCO 

10. Women of childbearing years must use adequate contraception (hormonal or 
barrier method of birth control) prior to enrollment 

5 11. Ability to understand and the willingness to sign a written informed consent 

document 

Exclusion Criteria: 

1, PAH associated with all other etiologies: HIV, portopulmonary disease, 
congenital heart disease (Humbert et al^ 2004) 

10 2. Subjects with pulmonary hypertension due to thromboembolism, significant 

interstitial lung disease, chronic obstmctive pulmonary disease, congestive heart failure, 
valvular heart disease (Humbert et aL, 2004) 

3. Subjects with (World Health Organization (WHO) functional Class IV (Humbert 
et al, 2004) 

15 4. Subjects with scleroderma with total limg capacity (TLC)< 60% of predicted 

within 30 days of screening 

5. Subjects with significant obstmctive lung disease with FEVl < 80% of predicted 

6. Subjects with hypotension defined as systolic arterial pressure < 90 xnmHg at 
baseline 

20 7. Subjects with hypertension defined as systolic arterial pressure >140 mmHg at 

baseline and a diastolic arterial pressure > 90 mmHg. 

8. Subjects with impaired renal function as defined as creatinine clearance <30 
ml/min as defined by the Cockcroft-Gault formula: Male: Creatitine clearance (ml/min)= 
(140-^age) X (body weight in kg)/ (72x serum creatinine in mg/dl); Female: Creatitine 

25 clearance (ml/min)= 0,85 (140-age) x (body weight in kg)/ (72x serum creatinine in mg/dl) 

9, Subjects with liver function tests (transaminases (AST/ALT), total bilimbin, and 
alkaline phosphatase) >2X normal values 

43 



wo 2007/087575 



PCT/US2007/060995 



10. Subjects with, acutely decompensated heart /faitoe or hospitalization within the 
previous 30 days prior to screening 

1 1 . Subjects may not be receiving any other investigational agents 

12. Subjects on endothelin receptor antagonists (bosentan, sitaxsentan^ ambrisentan) 
5 or chronic arginine supplementation 

13. Subjects with left ventricular ejection fraction <45% or left ventricular shortening 
fraction <0.2 

14. Subjects with acute myocardial infarction within 90 days prior to screening 

15. Subjects with limitations to performance of exercise measures (6MW) due to 
10 conditions other than PH associated dyspnea/fatigue 

16. Subjects taking nitrates for any medical problem 

17. Subjects taking phosphodiesterase inhibitors (any formulation) for erectile 
dysfunction 

18. Subjects with a recent (<180 days) history of pulmonary embolism verified by 
15 ventilation/perfiision scan, angiogram or spiral CT scan 

19. Pregnant or lactating women 

20. Subjects with a history of current drug abuse including alcohol 
Treatment Plan: 

Each subject will be individually dose escalated to a maximum of 400 mg twice daily. 

20 The starting dose of sorafenib will be 200 mg daily. If tolerated at the completion of 1 
month, the dose will be increased to 200 mg twice daily. If dose-limiting toxicity (DLT) 
occurs, dose escalation will be terminated and that dose level will be denoted as the 
maximum administered dose (MAD) for this subject. The subject will continue on the dose 
preceding the last escalation to complete a total of 4 months of active therapy. If this dose is 

25 not tolerated the subject will be withdrawn. If 200 mg twice daily is tolerated at the 
conclusion of month 2, the dose will be increased to 400 mg twice daily. If 400 mg twice 
daily is tolerated at the end of month 3 it will be continued until the completion of month 4. 



44 



wo 2007/087575 



PCT/US2007/060995 



Dose Limiting Toxicity (DLT) is defined as intolerable prostacyclin side effects: 
flushing, headache, nausea, loose stool, jaw discomfort with "first bite'\ and foot pain with 
prolonged standing or walking. Intolerable side effects from phosphodiesterase inhibitors: 
headache, gastrointestinal distress, hypotension. Intolerable side effects from sorafenib: rash^, 
5 diarrhea, fatigue, hypertension, hand-foot syndrome. 

Grading of DLT will occur on a 3 point scale for prostacyclin and phosphodiesterase 
side effects: 1 = stable, 2 = increased but tolerable, 3 == increased -f- intolerable. Prostacyclin 
dose will not be down-titrated for any reason in this investigational trial. If this is required 
the subject will be withdrawn. 

10 Grading of DLT for sorafenib will be based on the Common Temiinology for 

Adverse Event oncology grading scale 1-5. (CTAE V3. 0-1 2/1 2/03). The three most common 
adverse events include diarrhea and hand-foot skin reaction with the following grading: 
Diarrhea: 1 = increase of < 4 stools per day over baseline, 2 = increase of 4-6 stools per day 
over baseline; IV fluids indicated < 24 hours, 3 — increase of > 7 stools per day over baseline; 

1 5 incontinence; TV fluids >24 hours; hospitalization^ 4 = life threatening consequences, and 5 = 
death. Hand-foot skin reaction: 1 = minimal skin changes or dermatitis (e,g. erythema) 
without pain, 2 = skin changes (e,g. peeling, blisters, bleeding, edema) or pain, not interfering 
with function, and 3 ~ ulcerative dermatitis or skin changes with pain interfering with 
function. H3/pertension: 1 = asymptomatic, transient (<24 hours) increase by >20 nrniHg 

20 (diastolic) or to 150/100 mmHg if previously normal; intervention not indicated, 2 = recurrent 
or persistent (>24 hours) or symptomatic increase by >20 nmiHg (diastolic) or to >150/100 
mmHg if previously normal; monotherapy may be indicated, 3 = requiring more than one 
drug or more intensive therapy than previously, 4 = Life threatening consequences (e,g, 
hypertensive crisis), 5 ~ death. Subjects will be withdrawn with CTAE grade 3 diarrhea, 

25 hand-foot skin reaction, or hypertension. 

DLT Cardiovascular/PH toxicity is defined as arrhythmia, worsening right heart 
failure, hospitalization for worsening right heart failure/PH, worserdng dyspnea, worsening 
WHO class, worsening exercise capacity as defined as a decrease in 6MW >20% firom 
baseline or a decrease of >30 meters with subjective or clinical signs and symptoms of 
30 progression. 



45 



wo 2007/087575 



PCT/US2007/060995 



Expected Adverse Events (AE): Prostacyclin side effects: include flushing, headache, 
naxisea, loose stooU jaw discomfort with "first bite", and foot pain with prolonged standing or 
walking. Sildenafil side effects: gastrointestinal discomfort, headache, flushing. Sorafenib 
side effects: rash, diarrhea, fatigue, hypertension, hand-foot syndrome. 

5 Withdrawal: Subjects withdrawn from protocol due to clinical deterioration or DLT 

will have an office visit 30-40 days after end of treatment to record the following: physical 
exam, blood pressure, PH symptoms, WHO class, concomitant medications/adverse events, 
dose of prostacyclin, 6MW and TT. 

Data Safety Monitoring: Weekly meeting by the PH research team will discuss all 
10 subjects enrolled for data safety monitoring. 

Data Collection: Subjects will have a screening visit prior to enrollment including a 
history and physical exam. At this visit subjects will have two-6MW with Borg Dyspnea 
Score (B) >2 hrs apart up to 1 day later, as per ATS guidelines, using phrases of standard 
encouragement (ATS statement, 2002). The second test will be used for screening purposes 

15 and to limit variability. The mean reported increase ranges from 0 to 17% preformed a day 
later (ATS statement, 2002). Alternatively this can be done on separate days within 14 days. 
All patients will have an assessment of WHO functional class, and if needed PFT and a right 
heart catheterization as stated by the protocol. WHO functional Class is defined as: Class I: 
no limitation of physical activity, no symptoms of chest pain, angina, dyspnea, or near 

20 syncope with ordinary activity. Class 11: slight limitation of physical activity, ordinary 
physical activity causes dyspnea or fatigue, chest pain, or near-syncope. Class III: marked 
limitation of physical activity, less than ordinary activity causes dyspnea or fatigue, chest 
pain, or near-syncope, and Class IV: inability to perform activity without symptoms, signs of 
right heart failure, dyspnea and or fatigue at rest, and discomfort is increased by any physical 

25 activity (Humbert et al, 2004). These screening tests will be counted as their baseline test 
results. 

If the subjects meet criteria for enrollment they will return for the formal baseline visit 
approximately 14 days from screening. At this time they will have a 6MW/B followed by TT 
(unencouraged) >1 hr apart (Gomberg-Maitland et al, 2005; Patterson et al, 1972) a 2DE to 
30 assess TR velocity, a 3DE examination (experimental endpoint), and CT (experimental 
endpoint). Subjects will be seen weekly for safety evaluation with a clinical exam and 

46 



wo 2007/087575 



PCT/US2007/060995 



documentation of WHO classification. Safety evaluation will include the following data: 
adverse events related to sorafenib toxicity, related to prostacyclin side effects, and related to 
phosphodiesterase inhibitor side effects. Each month, all subjects will have both a 6MW and 
TT as per protocol^ laboratory including: CBC with differential, BCP (serum sodium), LFT 
5 (including albumin), uric acid, INR, BNP, troponin I, bFGF, and then be seen in the office to 
evaluate further dosing of sorafenib. 

At month 4 subjects will be required to be seen on 2 consecutive days. Day 1 subjects 
will have an office visit, a baseline 6MW/Borg score, an assessment of WHO fimctional 
class, a TT, PFT, 3DE, and CT. Day 2 will be a right heart catheterization. 

10 Hemodynamic values will be determined by serial measurements of hemodynamic 
parameters (specifically CO and mPAP) to demonstrate stability. Stable hemodynamics are 
defined by changes in CO and mPAP of less than or equal to 20% between three consecutive 
serial measurements at least 5 minutes apart. After hemodynamic stability is demonstrated, 
the hemodynamics and oxygen saturation variables from the last assessment will be recorded. 

15 Safety endpoints at month 4 will include: time to clinical worsening defined as death, 

lung transplantation, hospitalization due to PH/right heart failure, a decrease in exercise 
measures by at least 20% compared with baseline measure, worsening hemodynamics: either 
an increase in mean PA pressure by >20%, an increase in PVR > 20%, and or a decrease in 
CO by >20%, and worsening PFTs as evidence by a decrease in DLCO, FEVl, FVC, or TLC 

20 by >15%. Preliminary efficacy endpoints will include: monthly 6MW/B, WHO functional 
class, TT (experimental), 4 month right heart catheterization, TR velocity on 2DE, 3DE to 
assess RV parameters, (experimental)^ and a 64 slice CT (experimental). 

Follow-Up: At the conclusion of 4 months, subjects with perceived benefit as 
evidenced by objective and subjective measures based on the determination of the principal 
25 investigator and the sponsors and advisors on the grant will continue on therapy at their 
month 4 dose up to 1 year of therapy. They will continue to have follow-up visits every 3 
months with a 6MW test to a maximum of 1 year. They will be expected to speak with a 
member of the PH team to discuss AEs monthly by phone and all serious adverse events 
(SAE) will be reported to the PI and the IRB throughout the duration of the study. 

30 B. Single center Phase II trial of sorafenib in PAH patients. 



47 



wo 2007/087575 



PCT/US2007/060995 



Objectives Phase II study: Objectives are to assess safety and efficacy endpoints of 
sorafenib by a randomized discontinuation design. This is a 2 center study with a placebo 
controlled study using a randomized discontinuation design: placebo or MTD based on Phase 
IB study 

5 Accrual: Estimated 22-88 patients. Enrollment will be completed in 18-24 months— 

tliis will be based on preliminary efficacy firom Phase EB. 

Patient Selection: Eligibility and Exclusion Criteria as in Phase IB. 

Treatment Plan: 

Phase II will begin with a dose titration to the MTD based on the escalation described 
10 above; a 12-week open label run-in period. This will begin with a dose titration to the Phase 
IB MTD based on a titration regime of tolerance from the Phase IB study. If a subject does 
not tolerate any dose prior to the MTD or 1 month of MTD they will be withdrawn; not 
I'andomized into the placebo controlled study. Subjects with worsening 6MW > 20% with 
worsening fiinctional class or signs/symptoms of deterioration will be withdrawn firom the 
15 study. After the 12-week rua-in period, disease status will be assessed based on change in 
6MW distance. Subjects with > 50% improvement or > 100 meter improvement will 
continue to receive sorafenib until disease progression or toxicity, in order to avoid potential 
ethical concerns about randomization of patients with apparent major clinical benefit. The 
inventors do not expect a withdrawal for clinical deterioration with sorafenib but since this 
20 occurs with prostacyclin it will be a potential risk. These patients will be followed as per 
standard of care every 3-6 months. 

The remaining patients without obvious treatment benefit or failure during the open- 
label run-in period will be randomized in double-blind fashion using a central allocation via a 
telephone randomization system to receive the same dose of sorafenib or placebo. Evaluation 

25 of these subjects will be as per the Phase IB plan with weekly safety and monthly efficacy 
evaluations. Subjects with worsening 6MW > 20% with worsening fiinctional class or 
signs/symptoms of deterioration will be miblinded. If on placebo they will receive sorafenib 
at their previous dose; if on active drug they will be withdrawn. Those subjects who have a 
decline in 6MW without worsening clinical signs or symptoms will be seen at the next week 

30 visit and the 6MW will be repeated (unless there is evidence of clinical deterioration); this is 
to address the variability of the test and the variability of the individual. If the repeat 6MW 



48 



wo 2007/087575 



PCT/US2007/060995 



still demonstrates deterioration^ the subject will be imblinded to therapy and treatment will be 
as above: if on placebo they will receive sorafenib at their previous dose; if on active dn.ig 
they will be withdrawn. DLT criteria will be followed as per Phase IB at weekly visits. 

Data Collection: 

5 Subjects will be followed as per Phase IB with the following differences: 1) If 

subjects have clinical deterioration at any time after randomization, defined as 6MW > 20% 
decline with clinically assessed deterioration, or clinically assessed progression, they will be 
unblinded as per protocol. Subjects on placebo will be offered sorafenib at their previous 
dose, and subjects on active therapy, sorafenib will be withdrawn from study. 2) All subjects 
10 unblinded and started on active therapy will have a right heart catheterization after 6 months. 

"Withdrawal: 

Subjects withdrawn from protocol due to clinical deterioration or DLT will have an 
office visit 30-40 days after end of treatment to record the following: physical exam, blood 
pressure, PH symptoms, WHO functional class, concomitant medic ations/AEs, dose of 
15 prostacyclin, 6MW and TT. 

Statistical Analysis: 

Based on the efficacy data: improvement in * 6M W at 4 months on MTD a power 
calculation will be made to determine the sample size needed for this trial. The case report 
with combination therapy with imatinib at 3 and 6 months demonstrated a 60% improvement 

20 in 6MW. It is estimated that the sample size conservatively based on previous therapeutic 
trials to a maximum of 60% based on this report. A better estimate will be obtained based on 
Phase EB trial. For example, assuming a 30% improvement in 6MW5 with 80% power, the 
estimated sample size is 39 subjects in each arm, a 40% improvement 23 subjects in each 
arm, and a 50% improvement =15 subjects in each arm and a 60% improvement = 10 

25 subjects in arm. The investigators will allow for a 10% drop-out rate and adjust the sample 
size accordingly. An early stopping mle, using the O'Brien Fleming alpha spending rule, 
using an a of 0.0003 leaving p=0.0497 after 35% of subjects enroll (subject to change based 
on efficacy from IB). The trial will be stopped if the rate of deterioration at 3 months after 
randomization is higher or lower than expected. The DSMB will determine if the trial need 

30 to be stopped. 

49 



wo 2007/087575 



PCT/US2007/060995 



Data Safety Motiitoring Board: 

The DSMB will consist of the Sponsor, Co-Sponsor, members of the Scientific 
Advisory Committee in addition to 2 outside PH specialists to be determined by the Principal 
Investigator (PI). 

5 EXAMPLE 5 

Identification and initiation of early clinical trials of new agents in PAH 

The Clinical Therapeutics in Pharmacology and Pharmaco genomics group are 
informed of novel kinase inhibitors being evaluated by an oncology Phase I/II unit. As these 
agents are evaluated in oncology patients^ tlaey will be assessed for potential benefits and 
10 risks in PAH patients. If the agent scientifically has potential efficacy, the agent will be 
evaluated in preclinical studies with the hypoxic/SU5416 rat model of PAH. 

Studies will be designed as done with sorafenib, with 15-18 male Dahl-sensitive rats 
weighing between 150 and 250 grams will be divided into 5 groups: normoxia + vehicle 
(normal, healthy control), hypoxia + vehicle (hypoxia control), hypoxia + SU-5416 (positive 

15 control), hypoxia + sorafenib (safety comparison with SU-5416)^ and hypoxia + sorafenib + 
SU-541 6 (therapeutic activity assessment). All animal care and procedures will be preformed 
in accordance with institutional guidelines. Animals are housed in a Plexiglas chamber open 
to room air (normoxia) or maintained at 10% Fi02, Rats in the two SU-5416 groups will 
receive one injection of SU-5416 at the start of the experiment (20 mg/kg). Agent will be 

20 prepared for intraperitoneal lavage and dosed as per known rat protocols. Pressures and 
echocardiography will be preformed as described herein and in the scientific literature. If rats 
given the agent plus hj/poxia do not develop PAH and appear to have some therapeutic 
activity (i*ats given SU5416 plus hypoxia^ plus agent), the inventors will proceed to Phase IB 
study as described. 

25 Phase IB studies will be designed as a small safety and tolerability study of the agent. 

The study will determine the MTD and assess vital signs and clinical exam assessment at 
weekly office visits. If the drug is tolerated with some preliminary efficacy, a Phase 11 study 
will be initiated. The study will be a randomized discontinuation trial. If Phase I and Phase 
II are not successful because subjects do not tolerate the medication based on severe DLT, or 

30 with minimal efficacy the process will be reinitiated with a novel therapeutic agent. 



50 



wo 2007/087575 



PCT/US2007/060995 



EXAMPLE 6 

Additional Phenotvpic Markers 

Treadmill test as new standard of care exercise measure. The inventors have begun 
creating a new model using the data from the intravenous treprostinil study, based on the 
preliminary data demonstrating that the treadmill test (TT) was similar to the 6MW, had 
repeated reliability, and predicted functional class. The relationship between distance (DIST) 
and MET was modeled using generalized estimating equation (GEE) models which 
accoxmted for the within patients correlations since multiple measurements were conducted 
for each patient. The inventors first fitted a piecewise regression using data between baseline 
and month 3 (Model 1) and validated the model fitting using data between month 6 and 
month 12. The data had a ''good" correlation, a "good fit" and thus the inventors were able to 
develop a piecewise regression model with all data (Model 2). The new model appears to 
differentiate less sick patients and is as good as the 6MW, 

To completG analyses, the inventors will develop receiver operator curves accounting 
for repeated measures to compare the 6MW to the TT. Based on this analysis, if TT is as 
good as 6MW^ logistic regression analyses will be done on this data set to determine if MET 
is a good predictor of clinical worsening, survival, and clinical outcome variables including 
hemodynamics, echocardiography, and laboratory biomarkers. The model will estimate 
predicted MET based on 6MW distance but does not predict MET based on the patient's 
individual characteristics. At the University of Chicago, all patients have yearly TT 
evaluations, and evaluations pre and post medication changes. Initially, the model will be 
assessed in the University of Chicago dataset of patients having both the 6MW and the TT, 
A prediction equation will be developed for the TT to determine MET expected for individual 
PH patient based on WHO class. In addition, the inventors will be using the TT as an 
experimental endpoint in all trials conducted. 



51 



wo 2007/087575 



PCT/US2007/060995 



Table. 4 



Model 1: Model 2: 

Piecewise regression Piecewise regression 

Observed data (all with data 0-3 months with data 0-12 months 





— mnnthci 

1 1 1 1 LJI 1 LI 1 0 


^M" J ^ f / 




Mil' i } 








s.Iope <4 


107.1795 


slope <4 


110.3069 






slope ^ 


19.12348 


slope ^ 


19.37262 






intercept 


-31 .6585 


Intercept 


-42.90939 


MET 


mean DIST 


MET 


DIST 


MET 


DIST 


2 


176.0 


2 


183 


2 


178 


3 


288.4 


3 


290 


3 


288 


4 


395.8 


4 


397 


4 


398 


5 


415.1 


6 


416 


5 


418 


6 


442.2 


6 


435 


6 


437 


7 


460.1 


7 


454 


7 


456 


8 


476.9 


8 


474 


8 


476 


9 


423-5 


9 


493 


9 


495 


10 


513-8 


10 


512 


10 


515 


11 


545.5 


11 


531 


11 


534 



3DE and CT scans as novel non-invasive measures. Continued preliminary data will 
be obtained for both technologies. As development progresses, these measures will be linked 
5 with clinical phenotype obtained from the PHC database. Surprisingly on 3DE the tricuspid 
valve annulus does not dilate as expected with increasing RV size. Measures of RV 3D areas 
will be compared with 2D imaging using Spearman rank correlations. Change in area as a 
response to therapy will be recorded. TR velocity will be measured in 2DE. All of these 
measiues will be evaluated based on clinical characteristics, response to therapy, and 

10 outcomes to determine predictive value. Use of this modality in the evaluation of VEGF 
inhibitors and other vascular signaling targets on the right ventricle and pulmonaiy artery in 
our early drug development studies should prove to be a useful endpoint. For CT scans, the 
University of Chicago ''s 64 detector CT scan will be used to implement routine chest CT on 
all PH patients. The inventors contemplate using automated technology and advanced 

15 detection to identify plexiform lesions and novel indices of PH severity. The goal is to detect 
and then use the technology to follow response to therapy and as a possible screening tool. 

Biomarker development. Blood samples will be obtained from all patients seen by the 
PH program and stored for future evaluation of potential serum markers, genetic, and 
proteomic evaluation. Concurrent with Phase I/II trials, biomarkers previously described will 
20 be assessed: UFT (album.in)j uric acid, serum sodium, BNP, troponin I, bFGF, angiopoietin-2. 
Response to therapy may also be linked to genetic profiles. The GCRC will help with the 



52 



wo 2007/087575 



PCT/US2007/060995 



processing and storage of samples. A database has been created compliant with HIPPA 
standards. 

EXAMPLE 7 

Sorafenib Relieves Pulmonary Vressnre 

5 A3 week study was designed and executed to assess the safety and therapeutic 

activity of sorafenib in a hypoxia-induced model of pulmonary hypertension in Dahl SS rats. 
A compound with a reportedly more Hmited spectrum of kinase inhibitory activity- SU5416 
was previously demonstrated to exacerbate pulmonary hypertension in the Dahl rat 
hypoxemic model. As part of the spectrum of sorafenib pharmacologic activity overlaps 
10 with SU5416, we performed this study to determine whether sorafenib had similar effects or 
with additional mechanisms of inhibitory activity could counteract the effects of SU5416. 

Fifteen Dahl Salt Sensitive strain rats were divided into 5 groups: normoxia + vehicle 
(normal^ healthy control)^ hypoxia + vehicle (hypoxia control), hypoxia -f SU5416 (positive 
control), hypoxia + sorafenib (safety comparison with SU5416), and hypoxia 4- sorafenib + 

15 SU5416 (therapeutic activity assessment). Echocardiograms were performed on ail rats at the 
start of the experiment. Except for the normoxia group^ all rats were maintained in a hypoxia 
chamber with a partial pressure of oxygen of 10% Fi02 for the duration of the experiment. 
Rats in the 2 SU5416 groups received one subcutaneous injection of SU5416 at the start of 
the experiment (20 mg/kg). Stock sorafenib solutions were prepared every three days, 

20 crushing and dissolving sorafenib tablets in EL/ethanol (50:50; Sigma Cremophor EL, 95% 
ethyl alcohol) at final concentration of 4 mg / mL, protected trom light exposure and stored at 
room temperature. Final dosing solutions were prepared on the day of use by dilution of the 
stock solution to 1 mg/mL with water and administered by gavage to the rats daily. After 3 
weeks, all rats had echocardiography and hemodynamic studies. Organ and blood specimens 

25 were obtained for furtlier evaluation. 

As has been previously described, rats given SU5416 developed pulmonary 
hypertension measured by elevated right ventricular and pulmonary artery pressures, 
echocardiographic changes, and elevated right ventricle/Iefl ventricular weights. Rats 
exposed to h3^oxia had mildly elevated pressures compared with nomioxia and there was no 
30 significant change in pressures or weights in rats given hypoxia plus sorafenib. Sorafenib 
appears to have a beneficial effect on pulmonary hypertensive rats as rats in hypoxia, plus 

53 



wo 2007/087575 



PCT/US2007/060995 



SU5416, plus sorafenib had pressures and weights similar to normoxia. Table 4. The small 
sample size did not allow for significant change in echocardio graphic data but there appeared 
to be a positive trend with this combination. 

Sorafenib appears to protect SU5416 treated hypoxia-exposed rats jErom pulmonary 
5 arterial hypertension. Furthennore, while sorafenib and SU5416 share some pharmacological 
activity, unlike SU5416, sorafenib does not exacerbate hypoxia-induced hypertension. Based 
on these results the inventors repeated the experiment with 1 8 rats distributed in the same 5 
groups (only difference was 3 additional rats in the normoxia control group). The results were 
reproducible. In the initial experiment two rats died during induction of anesthesia (after 
10 unblinding normoxia , Hypoxia/SU5416). In the subsequent experiment, one rat died on day 
zero (after unblinding hypoxia/SU5416). 

Sorafenib appears to protect SU5416-treated hypoxia-exposed rats jGi-om pulmonary 
arterial hypertension. Furthermore, while sorafenib and SU5416 share some pharmacological 
activity, unlike SU5416 sorafenib does not exacerbate hypoxia-induced hypertension. 



54 



wo 2007/087575 



PCT/US2007/060995 



a 



o 







8.80±5.94 




6.80 


230i0.71 






5.63±4.07 






" 

vo 

CO 

41 

W-) 

oo 









CI (PA) 


to 

• 

1 — 1 

-H 
m 
est 

r<i 


CN 

ro 

CD 
-H 

T— * 

1 1 




1.16±0.30 


1.12±0.16 




1.21±0.26 


0.87±0.14 




1.70±1.43 


0.82±0.17 




0,94db0.49 


1.12±0.27 


CO (PA) 


736±478 


CD 
CO 

wo 




406±115 


oo 

oo 
<^ 

CO 




400±99 


279±69 




551±467 


271±48 




vo 
vo 

41 

CO 
CO 


vo 

CD 
1 — t 

ON 
WO 
CO 




ON 

rn 


8.75±0.64 




o 
cs 

CD 

wS 


4.43±1.78 




3.65±2.76 


CN 
0\ 

CD 

-H 
vo 

to 




CN 

WO 

» 

wo 

41 

CO 
1 — 1 

vd 


3.87±0,95 




2.00±1.18 


3.80±1.41 


MPA 


0.45±0.05 


0.43±0.05 




0.40±0.03 


0.40±0.03 




0.45±0.01 


0.41±0.05 




CO 

1 

OO 
CO 


0.38±0,07 




■1 — ( 

o 

41 
CO 


0.45±0.04 


CI (Ao) 


1.49i0.11 


1.00±0,09 




1.14±0.15 


0.91±0.35 




0.95±0.06 


CN 
CN 

* 

CD 

4H 

CN 

oo 

* 

<o 




o 

rO 

* 

1 — t 

4i 
t — 


1.00±0.80 




0.74±0.19 


WO 

vq 

<o 


CO (Ao) 


500±29 


CD 

^ 

OO 
CO 




397±36 


OO 
t — I 

CO 

CN 
CO 




310±10 


wo 
wo 

41 




oo 

^ H 

4H 

r- 
wn 


VO 

vo 

CN 
CO 




WO 
vo 

41 
t — 


vo 

CN 


LV Mass 
(gram) 


VO 
CD 

• 

CD 
WO 


CD 

OS 

CD 

• 




1.50±0.44 


CO 

1 — t 

CD 

-H 
oo 
o 

* 




m 

CD 
CO 

■ 


CO 
CO 

» 

CO 

CN 




CD 
CO 

cd 

"n 
O 
VO 


O 

y—i 
CD 

o 

CO 

* 




o 

4i 

CO 

• 


0.93±0.05 


FS (%) 


O) 

4H 
wo 


68±4 




79±3 


^ 1 




71±11 


CO 
vo 




WO 

oo 


o 

4i 
oo 

vo 




1 — I 
1 — ( 

41 


75±14 




0.24±0.06 


CN 
O 

CD 

di 

CN 




0.24±0.10 


O.I6±0,02 




CD 
C? 

vo 

1 — f 
CO 


0.16±0.02 




CO 

<o 
^[ 

4i 
1 — 1 

O 


0,18±0.03 




0.22±0.04 


0.20±0.04 


RVAW 


1 — 1 
CD 


T" ■! 

CD 






0.14 




0.07 


0.16±0.03 




t — t 
O 






0.09 


O.lliO.Ol 


Wt(g) 


337±19 


VO 
VO 

oo 

CO 




350il4 


353±25 




r — I 

41 

oo 
CN 

CO 


CN 

± 

CN 

CO 




o 

V— I 

oo 
CN 
CO 


331±15 




CO 

CO 
CO 


CO 

4? 
oo 

CO 




Nomioxia-PRE 

(n=3) 


Normoxia- 
POST (n=2) 




hypoxia-PRE 


hypoxia-POST 
(n=3) 




o 

CO ci 


it 

4- ^ — 

o 

CO cit 






H 

GO 

o 

*a 

<^ ^ 

O F=J 

CQ ■' 




wo 

>3 CO 

CO li 

S vo 


sorafemb+SU54 
16-POST (n=3) 



wo 2007/087575 



PCT/US2007/060995 



REFERENCES 

The following references, to the extent that they provide exemplary procedural or 
other details supplementary to those set forth herein, are specifically incorporated herein by 
reference. 

U.S. Patent 5,145,684 

U.S. Patent 5,399,363 

U.S. Patent 5,466,468 

U.S. Patent 5,543,158 

U.S. Patent 5,552,157 

U.S. Patent 5,565,213 

U.S. Patent 5,567,434 

U.S. Patent 5,580,579 

U.S. Patent 5,641,515 

U.S. Patent 5,641,515 

U.S. Patent 5,725,871 

U.S. Patent 5,738,868 

U.S. Patent 5,741,516 

U.S. Patent 5,756,353 

U.S. Patent 5,780,045 

U.S. Patent 5,792,451 

U.S. Patent 5,795,587 

U.S. Patent 5,804,212 

U.S. Patent Pub. 2001/0027202 

U.S. Patent Pub. 2003/0125359 

U.S. Patent Pub. 2003/0139605 



Allen and Chonn, FEES Lett,, 223(l):42-6, 1987. 

Arcasoy et al^ Am J Respir Crit Care Med 167:735-40, 2003 

ATS statement: guidelines for the six-minute walk test. Am J Respir Crit Care Med 166:1 1 1- 
7, 2002 

Badesch et al. Am. Coll CardioL, 43(12 Suppl S):56S-61S, 2004, 
Balasnbramaniam et ah. Am J Physiol Lung Cell Mol Yhysiol 284:L826-33, 2003 



56 



wo 2007/087575 



PCT/US2007/060995 



Baxst etaL, N EnglJ Med 334:296-301, 1996 
Barst, J Clin lavest 1 15:2691-4, 2005 

Benet et al.^ Goodman and Oilman's The Pharmacological Basis Of Therapeutics, Hardman 
et al (EdsO, 9^^ Ed., Chap, 1, McGraw-Hill, NY, 1996, 
5 Benisty et al. Chest 126:1255-61, 2004 

Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. 

Clinical Pharmacology and Therapeutics (CPT) 69:89-95, 2001 
Chandran et al, Indian J, Exp Biol, 35(8):801-809, 1997. 
Chu et al, Ann Thorac Surg 77:449-56, 2004 
1 0 Clapp et al , Am J Respir Cell Mol Biol 26 : 1 94-20 1 , 2002 
Cool etal. Am J Pathol 155:411-419, 1999 

Couvreur et al. Bull Mem. Acad. R. Med. Belg., 143(7-9):378-388, 1988. 

Couvreur et al, FEBSLett., 84(2):323-326, 1977. 

Couvreur et al, J. Pharm, Set, 69(2): 199-202, 1980. 
15 Couvretar, Crit, Rev. Ther, Drug Carrier Syst., 5(l):l-20, 1988. 

Cross et al. Trends Biochem Sci, 28:488-494, 2003. 

Czeslick et al^ Eur J Clin Invest 33:1013-7, 2003 

D^Alonzo et al. Aim Intern Med 1 15:343-9, 1991 

Data etal. Circulation 64:4-12, 1981 
20 Dorfinuller et al, Eur Respir J 22:358-63, 2003 

Douglas et al, Crit Rev Ther Drug Carrier Syst., 3(3):233-61, 1987, 

EddaMbi et al, J Clin Invest 2001 108:1 141-50, 2001 

Farber and Loscalso, N. Engl J. Med,, 351:1655-1665, 2004. 

Friede A, Grossman R, Hunt R, Li R, Stem S: National Biospecimen Network Blueprint, in 
25 Constella Group I (ed). Durham, NC, 2003 

Frost et al, Vascul Pharmacol 43:36-9, 2005 

Gabizon and Papahadjopoulos, Pmc. Natl Acad, Set USA, 85(18):6949-6953, 1988. 
Galie et al, N Engl J Med 353:2148-57, 2005 
Geraci et al, J Clin Invest 103:1509-15, 1999 
30 Ghofrani et al, J Am Coll Cardiol 42:158-64, 2003 
Ghofirani et al, N Engl J Med 353:1412-3, 2005 
Giaid and Saleh, New Engl J Med 333:214-21, 1995 
Giaid et al. New Engl J Med 328:1732-9, 1993 
Giaid, Chest 114:208S-212S, 1998 

57 



wo 2007/087575 



PCT/US2007/060995 



Giaxmelli et ah, Curr Med Res Opin 21:327-32, 2005 
Girgis et ah. Am J Physiol Heart Circ Physiol 285 :H93 8-45, 2003 
Girgis et ah. Am J Respir Grit Care Med 172:352-7, 2005 
Gomberg-Maitland et ah. Am J Respir Grit Care Med 172:1586-9, 2005 
5 Gomberg-Maitland et ah , Am J Cardiol 96: 1 334-6, 2005 
Henry-Michelland et ah. Int. J. Pharm., 35:121-127, 1987. 
Herve et ah. Am J Med 99:249-25 A, 1995 
Hirahatae/^a/., Gart To Kagaku Ryoho, 19(10):1591, 1992. 
Hoeper et ah, N Engl J Med 342:1866-70, 2000 
10 Homma et ah, J Heart Lung Transplant 20:833-9, 2001 
Hmnbert et ah, N Engl J Med 351:1425-36, 2004 

Hwang eria/., Crit Rev. Ther. Drug Carrier Syst., 15(3):243-284, 1998, 

Itoh et ah. Am J Respir Crit Care Med 169:34-8, 2004 

Kawut et ah Am J Cardiol 95: 199-203, 2005 
15 Kawut et ah. Chest 128:2355-62, 2005 

Kido et ah, J Thorac Cardiovasc Surg 129:268-76, 2005 

Kopec et ah, J Clin Epidemiol 46:959-71, 1993 

Kucher et ah , Arch Intem Med 165:1 777-8 1 ^ 2005 

Laliberte et ah, J Cardiovasc Pharmacol 44:209-14, 2004 
20 Lasic, Trends BiotechnoL, 16(7):307-321, 1998. 

Lee etah, J Clin Invest 101:927-934, 1998 

Leuchte et ah. Chest 128:2368-74, 2005 

Leuchte et ah, J Am Coll Cardiol 43:764-70, 2004 

Marcos et ah, Circ Res 94:1263-70, 2004 
25 Margalit, Crit Rev. Ther. Drug Carrier Syst, 12(2-3):233-261, 1995. 

Mathiowitz etah, Nature, 386(6623):410-414, 1997. 

McLaughlin et ah. Circulation 106:1477-82, 2002 

McLaughlin etah. Chest, 126(1):78S-92S, 2004. 

Morishita et ah, Proc, Nath Acad, ScL USA, 90:8474, 1993. 
30 Nagaya et ah. Am J Respir Crit Care Med 1 60:487-492, 1999 

Nagaya et ah. Circulation 102:2005-10, 2000 

Nauser and Stites, Am, Fam. Physician., 63(9): 1789-1798, 2001. 

Olschewski et ah, N Engl J Med 347:322-9, 2002 

Gudiz and Ginzton, J Heart Lung Transplant 22:832-3, 2003 



58 



wo 2007/087575 



PCT/US2007/060995 



Oudiz et aL, Am J Cardiol 97:123-126, 2006 

Park et al. Congest Heart Fail 10:221-5, 2004 

Patterson et al^ Am J Cardiol 30:757-62, 1972 

Petrova et al Exp Cell Res, 253:117-130, 1999 
5 Physician's Desk Reference 2003. 

Pinto-Alphandary et al, X Drug Target, 3(2):167-169, 1995. 

Quintanar-Guerrero et al, Pharm, Res., 15(7):1056-1062, 1998. 

Remington's Pharmaceutical Sciences" 18*^ Ed., 1545-1568, 1990. 

Remington's Pharmaceutical Sciences" 18^^ Ed,, 1596-1609, 1990. 
10 Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 711, 1990. 

'Rioh^ Advances in Ptdmonary HypertensioTi , 1(1):3, 2002. 

Rich, Ann, Intern. Med., 1 07(2) :2 16-223, 1987. 

Rondelet et al. Circulation 1 10:2220-5, 2004 

Rosner et al, J Clin Oncol 20:4478-84, 2002 
1 5 Rubin et al , Expert Opinion 1 1 :991 -1 002, 2002 

Rubin, Advances in Pulmonary Hypertension, 1(1): 16 and 19, 2002 

Rubin, Chest, 126(1):4S-6S; 7S-10S, 2004. 

Rubin, Tfie New England Journal of Medicine, 33 6(2): 111, 1 997 . 
Schermuly et al, J Clin Invest 1 15:2811-21, 2005 
20 Simonneau et al. Am J Respir Crit Care Med 165:800-4, 2002 
Sitbon et al, .J Am Coll Cardiol 40:780-8, 2002 

Somiari et al. Transactions of the Integrated Biomedical Informatics & Enabling 
Teclmologies Symposium Journal 1:131-143, 2004 

Stadler et al, 1 Clin Oncol 23:3726-32, 2005 
25 Stella et al , In: Prodrugs: A Chemical Approach to Targeted Drug Delivery, Borchardt et al 

(Ed.), Humana Press, 247-267, 1985. 

Stiebellehner et al. Chest 123:1293-5, 2003 

Strumberg et al, J Clin Oncol 23:965-72, 2005 

Sullivan et al, Proc Natl Acad Sci U S A 100:12331-12336, 2003 
30 Takakura, Nippon Rinsho, 56(3):691-695, 1998, 

Takenaga J, Control Release, 52(l-2):81-87, 1998. 

T^cpson, Advances in Ptdmonary Hypertension, 1(1):16-17, 2002 

Taraseviciene-Stewart et al, FASEB J 15:427-38, 2001 

The Merck Manual, 17*^ Ed., 595, 1999. 



wo 2007/087575 



PCT/US2007/060995 



Torbicld et ah. Circulation 108:844-8, 2003 

Tuder et ah. Am J Pathol 144:275-285, 1994 

Tuder et al. Am J Respir Crit Care Med 159:1925-1932, 1999 

Tuder et al, J Paiiiol 195:367-74, 2001 

Tuder, Semin Respir Crit Care Med 15:207-214, 1994 

Ushio-Fukai et al, Arterioscler Thromb Vase Biol 21:489-95, 2001 

Wade et al, J Clin Pharmacol 44:83-8, 2004 

Wagenvoort and Mulder, Chest: 844-849, 1993 

Wagenvoort aad Wagenvoort, Pathology of pulmonary hypertension (ed 2nd). New York, 

John Wiley and Sons, 1977 
Welshed a/.. Chest 110:710-717, 1996 
Wilhelm et ah. Cancer Res 64:7099-109, 2004 

Wilman, In: Prodrugs in Cancer Chemotherapy, Biochemical Society Transactions, 14:375- 

382, Belfast, 1986. 
Xin et aL, Biochem Biophys Res Commun 204:557-64, 1994 
Yasunobu et al. Chest 127:1637-46, 2005 
Zambaux et al J, Protein Cheim, 17(3):279-284, 1998. 
zur Muhlene^ al, Eur. J. Pharm. Biopharm., 45(2): 149-1 55, 1998. 



60 



wo 2007/087575 



PCT/US2007/060995 



CLAIMS 

1 . A method comprising providing sorafenib to a subject with pulmonary arterial 
hypertension (PAH), with symptoms of PAH, or at risk for PAH. 

2. The method of claim 1, wherein the subject is provided with sorafenib by 
administering or prescribing to the subject sorafenib, a sorafenib prodmg, or a 
pharmaceutically acceptable salt thereof. 

3. The metliod of claim 1 ^ fiortlier comprising identifying a subject with PAH or 
symptoms of PAH. 

4. The method of claim 3^ wherein the subject is diagnosed as having severe PAH. 

5. The method of claim 1^ wherein the patient has a mean pulmonary artery pressure 
equal to or greater than 25 mm Hg with a pulmonary capillary or left atrial pressure equal to 
or less than 15 mm Hg, 

6. The method of claim 1, further comprising evaluating PAH in the subject. 

7. The method of claim 6, wherein the patient is evaluated before and/or after 
administering the composition. 

8. The method of claim 6, wherein the subject is evaluated for PAH by having an 
electrocardiogram, an echocardiogram, pulmonary function tests (PFTs), a perfusion lung 
scan^ and/or a right-heart cardiac catheterization. 

9. The method of claim 1, wherein the subject is provided multiple doses of sorafenib. 

10. The method of claim 9, wherein a dose is between about 5 and about 500 mg of 
sorafenib or a sorafenib prodrug. 



61 



wo 2007/087575 



PCT/US2007/060995 



1 1 , The method of claim 2, wherein the sorafenib or sorafenib prodrug is administered or 
prescribed for administration orally, intravenously, intraarterially, inhalation. 



12. The method of claim 1, further comprising providing at least a second PAH treatment. 



13. The method of claim 12, wherein the second treatment is an anticoagulant, an calcium 
chaimel blocker, a prostacyclin, Bosentan, nitric oxide. Sildenafil, a diuretic, a cardiac 
glycoside, a vasodilator, an endothelin antagonist, a phosphodiesterase inhibitor, an 
endopeptidase inhibitor, a lipid lowering agent, a thromboxane inhibitor, or oxygen. 



14. The method of claim 1, wherein the sorafenib is provided before, after, or during 
surgery. 



1 5. The method of claim 14, wherein the surgery comprises lung transplantation. 



16. A method of reducing pressure in the puhnonary artery of a patient comprising 
administering to the patient an effective amount of a composition comprising sorafenib, a 
sorafenib prodmg, or a pharmaceutically acceptable salt thereof 



17. A method for treating or preventing PAH in a patient comprising administering to the 
patient an effective amount of sorafenib, a sorafenib prodmg, or a pharmaceutically 
acceptable salt thereof. 



18. A pharmaceutical composition comprising (a) sorafenib or a sorafenib prodrug and 
(b) at least a second PAH treatment. 



19- The pharmaceutical composition of claim 18, wherein the second PAH treatment 
comprises an anticoagulant, an calcium channel blocker, a prostacyclin, Bosentan, nitric 
oxide, Sildanefil, a diuretic, a cardiac glycoside, a vasodilator, an endothelin antagonist, a 



62 



wo 2007/087575 



PCT/US2007/060995 



phosphodiesterase inhibitor, an endopeptidase inMbitor, a lipid lowering agent, or a 
thromboxane inhibitor. 

20. A method comprising providing a VEGFR2 inhibitor to a subject with pulmonary 
arterial hypertension (PAH), with symptoms of PAH, or at risk for PAH. 

21 . The method of claim 20, wherein the VEGFR2 inhibitor is selected from abt-869, 
amg706, AZD217U bay57-9352, sorafenib, XL647, XL999, GW786034, bevacizumab, 
PKC412, AJEE788, PTK787 (vatalanib), OSI-930, OSI-817, SU11248, AG-013736, ZK3- 
4709, quinazoline ZD6474, pyrrolocarbazole CEP-7055, or CP-547632. 



63 



wo 2007/087575 



1/17 



PCT/US2007/060995 



40 



35 



30 



20 



15 



10 



0 




4®i 



Mi 





fell' 



H Normoxia 



H Hypoxia 



Q Hypoxia + 
SU5416 



Hypoxia + 
SUS416 + 
Sorafenib 

Hypoxia + 
Sorafenib 



.J 



FIG. lA 



wo 2007/087575 



2/17 



PCT/US2007/060995 





45 




40 - 




■35 






Z3nii 


30 








25 - 


< 


20 - 






15 - 




10 - 




5 - 




0 ■ 



Pulmonary Artenai Pressures 



Vehicle 




I 



-',''•7-" 




Sorafenib 



^p<0.01;n=6 




Norm Hypoxia Hypoxia Hypoxia 

+ SU5416 



Hypoxia 
+ SU5416 



FIG. IB 



wo 2007/087575 



3/17 



PCT/US2007/060995 



E 
E 



40 



35 - 



30 



2^ 



20 



o: 15 



10 



0 




m Normoxia 



□ 



Hypoxia 



D Hypoxia + 
SU5416 



Hypoxia + 
SU5416 + 

Sorafenib 

Hypoxia + 
Sorafenib 



FIG. 2 



wo 2007/087575 



4/17 



PCT/US2007/060995 




50 
45 
40 
35 
30 

20 
15 
10 
5 
0 



■Jr-- 




^^^^^ 




I 



lii 






H Normoxia 




Hypoxia 



n Hypoxia + 
SU5416 



□ Hypoxia + 
SU5416 + 
Sorafenib 




Hypoxia + 
Sorafenib 



FIG. 3 



wo 2007/087575 PCT/US2007/060995 

5/17 



.2 0.3 



CO 

+ 



0.45 



0.4 



0.35 



0.25 



0.2 



0.15 



0.1 



0.05 



0 



Hi 






Normoxia 



Hypoxia 



□ Hypoxia + 
SU5416 

□ Hypoxia + 
SU5416 + 
Sorafenib 

Hypoxia + 
Sorafenib 



FIG. 4A 



wo 2007/087575 



6/17 



PCT/US2007/060995 



Right Ventricular Hypertrophy 



CD 

CO 



0.5 ^ 
0.45 

0.4 
0.35 J 

0.3 - 
0.25 _ 

0.2 . 
0.15 - 

0.1 - 
0.05 



^p<0.01 




p<0-01 
Sorafenib 



1 




Normoxia 



Hypoxia Hypoxia + Hypoxia Hypoxia + 

SU5416 SU5416 



FIG. 4B 



wo 2007/087575 



7/17 



PCT/US2007/060995 



140 



120 



100 



80 



40 



20 



0 



60 -i 




"^.;j;,-;v;jVrf ij.^ 

■'"■•3, 



in Normoxia 



Hypoxia 



n Hypoxia + 
SU5416 

□ Hypoxia + 
SU5416 + 
Sprafenib 

Hypoxia + 
Sorafenib 



1 



FIG. 5 



wo 2007/087575 



8/17 



PCT/US2007/060995 



LV Mass (g) 



2,50 



2.0D 



1.50 



1.00 



0.5O 



Q.0O 




0mm 




I 



'i?" -.-fit' , 






ills 




^ ^*C*^rs;.Tf *^ ; 



ft -■rSM-'j' 




m 




NML- NML- 
pre post 



HYP- HYP- 
pre post 



HYP HYP 

+ + 

SOR- SOR- 

pre post 



HYP HYP 

+ + 

SU5416 - 
pre posi 



HYP HYP 
+ 4, 

SOR SOR 

SU5416 
pre post 



L 



FIG. 6 



wo 2007/087575 



9/17 



PCT/US2007/060995 



Aortic Cardiac Output (l/min) 



1200.00 



1000.CX) 



800.00 



600.00 



400.00 




200.00 - 



0,00 



Jit J ^r^ 'trinJ 







'*^*-i^v-i*j:'i 

.V- 



--^irf*'"^*--" -'i^ii V;5 ;'iii J 



NML- NML- 
pre post 



HYP- HYP- 
pre post 



HYP HYP 

SOR- SOR- 
pr© post 



HYP HYP 

SU5416 
pre post 



HYP 

4- 



HYP 



SOR SOR 

SUS416 
pre post 



J 



FIG. 7 



wo 2007/087575 



10/17 



PCT/US2007/060995 



PA Pressure Gradient (mm Hg) 



14.00 



12.00 



1O.00 



8,00 



6.00 



4.00 




2.00 



0.00 




^-J'^ 




d. ■-is;.;^,^,;^t^,■ 



. '-^ 



0,!-it-,(-i; „ :- 




NML- NML- 
pre post 



HYP- HYP- 
pre post 



HYP + HYP + 
SOR- SOR- 
pre post 



HYP 4- HYP + 



SU541S 



HYP + HYP + 
SOR SOR 

SU5416 
pre post 



FIG. 8 



wo 2007/087575 



11/17 



PCT/US2007/060995 



RV free wall thickness (cm) 



0.2 1 



o.ia 



0.16 



0.14 



0.1 



o.oa 



G.06 




0.O4 . 



ao2 




.lit);. -jriiT, 






; | ,^.^; i ^tj.; 







.^.mi H ji* 1^^. 












mi 





'IS 

^ ^-^^jf 









• ' ' ^^^^^^♦^^.•'Vji^ ^^^^^^^^ 



iJ,,,(!J,.._t>. 



i-v-i.'iSJr.vii.-*; 



ftij^jj 





NML- NML- 
pre post 



HYP- HYP- 
pre post 



HYP + HYP + 
SOR- SOR- 
pre post 



HYP + HYP + 

SU5416 • 
pre post 



HYP + HYP + 
SOR SOR 

+ + 
SU6416 

pre post 



FIG. 9 



wo 2007/087575 



12/17 



PCT/US2007/060995 



PA Pressure (mm Hg) 



14.00 



12.00 



10.00 



8,00 



6.00 



4.00 



ZOO 



0.00 1^ 




NML-post 



NML-post 



Hyp+ 

SUS4^6pre 



Hyp+Sor+ 
SU5416 post 



Hyp i-Sor+ 
SU5416 pre 



IIyp+Sor+ 
SU5416 post 



FIG. 10 



wo 2007/087575 



13/17 



PCT/US2007/060995 



RV thickness (cm) 



Q,2 



ai8 



OAS 



0.14 




ai2 



O.00 



O.06 



0.04 



O02 



. ,iS#'*^3JT.^,1'-•■^,^i^i -, l>i ■ii 












NML-post NML-posl 



Hyp+ 

SU5416Dre 



Hyp+Sor+ 
SU5416 DOSt 



Hyp+Sor-f- 
SU5416 pre 



Hyp+Sor+ 
SU5416post 



FIG. 11 



wo 2007/087575 



14/17 



PCT/US2007/060995 




FIGs. 12A-12B 




FIG. 13 



wo 2007/087575 



15/17 



PCT/US2007/060995 





Imatinib ("2O0 mg/da)') 



Combination !tlierap<>''witli iloprost. 
sildenafil, and bosenitari 




FIG. 14 




FIG. 15 



wo 2007/087575 



16/17 



PCT/US2007/060995 




17/17 



PCT/US2007/060995 





3 
CO 






+ 






LL. 






CO 


CO 


+ 


+ 


+ 


ZZZ! ajEZ 


X 


X 












9 opBJO 
9 epejo 

Z epB4o 
I, epejo 
t/O apejo 

0 ©PBJQ 



UD ^ CO CN 

sieuiiuv # 



^^^^^